Cognitive Dysfunction and Diabetes Mellitus: Christopher T. Kodl and Elizabeth R. Seaquist

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

0163-769X/08/$20.

00/0 Endocrine Reviews 29(4):494 –511


Printed in U.S.A. Copyright © 2008 by The Endocrine Society
doi: 10.1210/er.2007-0034

Cognitive Dysfunction and Diabetes Mellitus


Christopher T. Kodl and Elizabeth R. Seaquist
Department of Medicine, Division of Endocrinology and Metabolism, University of Minnesota, Minneapolis, Minnesota
55455

The deleterious effects of diabetes mellitus on the retinal, significant roles. Modalities to study the effect of diabetes on
renal, cardiovascular, and peripheral nervous systems are the brain have evolved over the years, including neurocogni-
widely acknowledged. Less attention has been given to the tive testing, evoked response potentials, and magnetic reso-
effect of diabetes on cognitive function. Both type 1 and type 2 nance imaging. Although much insightful research has exam-
diabetes mellitus have been associated with reduced per- ined cognitive dysfunction in patients with diabetes, more
formance on numerous domains of cognitive function. The needs to be understood about the mechanisms and natural
exact pathophysiology of cognitive dysfunction in diabetes is history of this complication in order to develop strategies for
not completely understood, but it is likely that hyperglycemia, prevention and treatment. (Endocrine Reviews 29: 494 –511,
vascular disease, hypoglycemia, and insulin resistance play 2008)

I. Introduction nitive dysfunction, and many patients fear that recurrent


II. Cognitive Dysfunction in Patients with Diabetes hypoglycemia will impair their memory over time. Although
A. Type 1 diabetes much research has been done, the pathophysiology under-
B. Type 2 diabetes lying this complication is not well understood, and the most
C. Hypoglycemia and cognitive dysfunction appropriate methods to diagnose, treat, and prevent cogni-
D. Section summary tive dysfunction in diabetes have not yet been defined. In this
III. Pathophysiology of Cognitive Dysfunction in Diabetes article, we will review the nature of cognitive dysfunction in
A. The role of hyperglycemia type 1 and type 2 diabetes mellitus, the pathophysiology of
B. The role of vascular disease cognitive dysfunction secondary to diabetes, methodologies
C. The role of hypoglycemia
used to assess cognitive deficits in patients with diabetes, and
D. The role of insulin resistance and amyloid
potential future directions of research that are needed to
IV. Modalities for Assessment of Cognitive Dysfunction in
Patients with Diabetes
advance our understanding of this often overlooked com-
V. Future Directions plication of diabetes.
VI. Conclusion The purpose of this article is to present a comprehensive
review of the literature regarding the subject of cognitive
dysfunction in diabetes mellitus. To do this, we performed
I. Introduction MEDLINE searches for such key words and terms as “diabetes
mellitus,” “cognitive function,” “cognition,” “hypoglycemia,”
D IABETES MELLITUS IS a complex metabolic disease
that can have devastating effects on multiple organs
in the body. Diabetes is the leading cause of end stage renal
“insulin resistance,” and “Alzheimer’s disease,” among others.
We then pursued articles referenced in these sources. Although
this is a comprehensive review, it is not exhaustive. In addition,
disease in the United States (1) and is also a common cause
it should be noted that the field of cognitive dysfunction in
of vision loss, neuropathy, and cardiovascular disease. A less
diabetes is still in its early stages. It must be remembered that
addressed and not as well recognized complication of dia-
although there have been many significant contributions re-
betes is cognitive dysfunction. Patients with type 1 and type
garding the association of diabetes and cognitive dysfunction
2 diabetes mellitus have been found to have cognitive deficits
and many hypotheses based on this association, the causative
that can be attributed to their disease. Both hypoglycemia
mechanisms of diabetes on cognitive dysfunction are still un-
and hyperglycemia have been implicated as causes of cog-
dergoing development.

First Published Online April 24, 2008


Abbreviations: AGE, Advanced glycation end product; APOE-␧4, II. Cognitive Dysfunction in Patients with Diabetes
apolipoprotein E type 4; APP, amyloid precursor protein; DCCT, Dia-
betes Control and Complications Trial; EEG, electroencephalograph(y); A. Type 1 diabetes
fMRI, functional MRI; GLUT, glucose transporter; HbA1c, glycated he-
moglobin; MRI, magnetic resonance imaging; NMDA, N-methyl-d- Cognitive dysfunction in patients with diabetes mellitus
aspartate; PET, positron emission tomography; PPAR-␥, peroxisome
was first noted in 1922, when patients with diabetes, who
proliferator-activated receptor-␥; SPECT, single photon emission com-
puted tomography. were “free from acidosis but usually not sugar free,” were
Endocrine Reviews is published by The Endocrine Society (http:// noted to have impaired memory and attention on cognitive
www.endo-society.org), the foremost professional society serving the testing compared with controls (2). Since then, there have
endocrine community. been many studies designed to better delineate the scope and

494

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 22 May 2014. at 00:09 For personal use only. No other uses without permission. . All rights reserved.
Kodl and Seaquist • Cognition and Diabetes Mellitus Endocrine Reviews, June 2008, 29(4):494 –511 495

TABLE 1. Summary of cognitive domains that have been found to tions, but not with glycemic control in these populations.
be negatively affected by type 1 diabetes mellitus However, this last finding was confounded by the hetero-
geneity of how different studies defined “well” vs. “poorly”
Slowing of information processing*
Psychomotor efficiency* controlled diabetes (22).
Attention* As was demonstrated in the work of Brands et al. (22),
Memory cognitive function may be worse in patients with type 1
Learning diabetes who experience other diabetes complications. Def-
Problem solving
Motor speed icits in fluid intelligence, information processing, attention,
Vocabulary and concentration have been associated with the presence of
General intelligence background retinopathy (23). Proliferative retinopathy, ma-
Visuoconstruction* crovascular complications, hypertension, and duration of
Visual perception
diabetes were associated with poorer performance on tests
Somatosensory examination
Motor strength measuring psychomotor speed and visuoconstruction ability
Mental flexibility* in patients with type 1 diabetes (4 – 6). Patients with distal
Executive function symmetrical polyneuropathy displayed worse cognitive
Domains marked by asterisks have particularly strong support- function on most cognitive domains except for memory (5).
ing data. However, other studies were unable to identify a relation-
ship between impaired cognitive function and diabetic com-
magnitude of cognitive dysfunction in diabetes (Table 1). The plications (24). Future study will be necessary to determine
most common cognitive deficits identified in patients with whether there is a link between complications and alterations
type 1 diabetes are slowing of information processing speed in cognition.
(3– 6) and worsening psychomotor efficiency (3, 4, 7). How- Although complications like retinopathy and nephropa-
ever, other deficits have been noted, including deficits in thy usually require years of diabetes before becoming clin-
motor speed (5, 8 –10), vocabulary (7, 11–13), general intel- ically apparent, the onset of cognitive impairment has been
ligence (12, 14), visuoconstruction (6, 12), attention (6), so- found to occur early in patients with type 1 diabetes. Deficits
matosensory examination, motor strength (10), memory (7), in cognitive function have been detected as early as 2 yr after
and executive function (7, 14). Glycemic control appears to diagnosis in children with type 1 diabetes, and these patients
play a role in cognitive performance in patients with type 1 experienced less positive changes than controls over time in
diabetes. Functions such as psychomotor efficiency, motor general intelligence, vocabulary, block design, speed of pro-
speed (5, 15), attention, verbal IQ scores (16 –18), memory, cessing, and learning (12). Six years after diagnosis, these
and academic achievement (17) are improved with better same subjects had impaired IQ, attention, processing speed,
glycemic control. Specifically, an 18-yr follow-up of the Di- long-term memory, and executive function compared with
abetes Control and Complications Trial (DCCT) showed that controls (14). The age of onset of type 1 diabetes may also
those patients with type 1 diabetes mellitus with a time contribute to the presence of cognitive dysfunction, because
weighted mean glycated hemoglobin (HbA1c) less than 7.4% those who developed type 1 diabetes at less than 4 yr of age
performed significantly better on tests of motor speed and had impaired executive skills, attention, and processing
psychomotor efficiency than those subjects whose time speed when compared with those that were diagnosed after
weighted mean HbAlc was greater than 8.8% (15). In addi- 4 yr of age (14). Of note, chronic disease and time away from
tion, slowing of all cognitive function, an increased number school (secondary to illness, etc.) were not controlled for in
of mental subtraction errors (19), loss of inhibition and focus these studies.
(20), impaired speed of information processing, decreased Interestingly, several studies have shown patient gender
attention, and impaired working memory (21) have all been to influence neurocognitive function in patients with type 1
noted during acute hyperglycemia in patients with type 1 diabetes mellitus. Skenazy and Bigler (10) found that men
and type 2 diabetes. with type 1 diabetes had reduced performance on oscillation,
A recent meta-analysis included 33 studies examining cog- strength grip, and somatosensory testing compared with
nitive function in adult subjects with type 1 diabetes mellitus male controls, and the magnitude of this difference was
(22). It found that there were significant reductions in overall greater than that measured between women with type 1
cognition, fluid and crystallized intelligence, speed of infor- diabetes and their gender-matched controls. In addition, a
mation processing, psychomotor efficiency, visual and sus- decline in verbal intelligence was seen in boys with type 1
tained attention, mental flexibility, and visual perception in diabetes between the ages of 7 and 16, which correlated with
subjects with type 1 diabetes compared with controls. There worse glycemic control. This was not seen in girls of similar
was no difference in memory, motor speed, selective atten- ages (13). However, most human studies have not distin-
tion, and language. All studies included healthy matched guished between genders when describing results of neu-
control groups and used reliable testing measures at normal rocognitive testing, and therefore more controlled analysis
blood glucose values. Most studies included in the meta- should be done before any conclusions are drawn.
analysis controlled for depression; however, similar findings Of note, the strength of these neurocognitive studies is
were seen in those studies that did not control for depression. variable. Covariates that could affect neurocognitive testing
It is unclear whether any of these studies controlled for other include age, education, sex, history of other chronic illnesses,
chronic diseases that could affect cognitive function. Worse psychiatric disorders, neurological disorders, substance
cognition was associated with increased diabetes complica- abuse, absence from school, socioeconomic status, and hy-

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 22 May 2014. at 00:09 For personal use only. No other uses without permission. . All rights reserved.
496 Endocrine Reviews, June 2008, 29(4):494 –511 Kodl and Seaquist • Cognition and Diabetes Mellitus

poglycemia/hyperglycemia during testing. The reviewed re- served (46). In a population of nearly 2000 postmenopausal
ports controlled for at least some of these covariates, however women, Yaffe et al. (47) found that those with a HbA1c of
most fail to control for all of them. For example, only two more than 7.0% had a 4-fold increase in developing mild
studies that have been mentioned (6, 24) have reported con- cognitive impairment. Grodstein et al. (30) found that elderly
trolling for hyperglycemia at the time of testing, which has subjects who took oral diabetic medication, unlike those on
been proven to affect cognitive function (see Section II.A). The insulin, had similar scores on general tests for cognition as
cognitive domains that are affected by type 1 diabetes with subjects without diabetes. Other studies have demonstrated
the best evidence based on our review are indicated in Table 1 an inverse relationship between HbAlc and working mem-
with an asterisk. ory (27, 28), executive functioning (27), learning (26), and
complex psychomotor performance (26, 48) in patients with
B. Type 2 diabetes type 2 diabetes mellitus, supporting the hypothesis that
worsening glucose control leads to worsening cognitive
Patients with type 2 diabetes mellitus have also been function much like with type 1 diabetes. Also similar to type
found to have cognitive impairment (Table 2). Type 2 dia- 1 diabetes is the association between alterations in cognitive
betes has been associated with decreases in psychomotor function in patients with type 2 diabetes and diabetes com-
speed (25, 26), frontal lobe/executive function (26–28), verbal plications like peripheral neuropathy (28) and duration of
memory (29), processing speed (29), complex motor func- type 2 diabetes (25, 33).
tioning (26), working memory (27, 28), immediate recall, Impaired glucose tolerance without diabetes is also a risk
delayed recall (30), verbal fluency (26, 31), visual retention factor for cognitive dysfunction. Multiple investigations of
(32), and attention (33). The impact of these subtle neuro- patients with impaired glucose tolerance have shown them
cognitive deficits on the daily lives of patients with type 2 to have lower mini-mental status exam and long-term
diabetes is not clear. Sinclair et al. (34) found that subjects memory scores (49), impaired verbal fluency (31), increased
with mini-mental status exam scores less than 23 fared worse Alzheimer’s dementia (39), and increased vascular dementia
on measures of self care and ability to perform activities of (38) compared with control subjects. These observations mir-
daily living. These subjects also displayed an increased need ror the positive relationship found between hyperglycemia
for personal care and increased rates of hospitalization when in patients without diabetes and cardiovascular disease
compared with controls. Patients with diabetes also have (50 –52). The pathophysiology of this relationship is unclear,
been found to have slower walking speed, lack of balance, and there is evidence that both hyperglycemia and other
and increased falls associated with type 2 diabetes, but aspects of insulin resistance could contribute to this, which
whether the cerebral affects of diabetes contributed to these will be addressed later. Of note, however, not all studies found
abnormalities is debatable (35). Complicating the impact of that patients with impaired glucose tolerance (33, 53, 54) or type
mild neurocognitive dysfunction secondary to diabetes on 2 diabetes mellitus (54, 55) perform worse than normoglycemic
daily living is the observation that patients with diabetes are individuals.
twice as likely to have depression (27, 36), which will also However, like neurocognitive studies examining type 1
negatively affect cognitive function and daily activities. Type diabetes, the strength of these neurocognitive studies eval-
2 patients also have an increased incidence of Alzheimer’s uating type 2 diabetes and impaired glucose tolerance is
disease (37– 44) and increased incidence of vascular dementia variable. Although most of these studies controlled for
(38, 42, 45). Recently, Bruce et al. (36) found that 17.5% of age, there was uneven control for other covariates includ-
elderly patients with type 2 diabetes had moderate to severe ing education, psychiatric disorders, neurological disorders,
deficits in activities of daily living, 11.3% had cognitive im- hyperglycemia and hypoglycemia during testing, and chronic
pairment, and 14.2% had depression. illness. The cognitive domains that are affected by type 2 dia-
Glycemic control appears to play a role in determining the betes with the best evidence based on our review are indicated
degree of cognitive dysfunction detected in patients with in Table 1 with an asterisk.
type 2 diabetes, although this has not uniformly been ob-

TABLE 2. Summary of cognitive domains that have been found to C. Hypoglycemia and cognitive dysfunction
be negatively affected by type 2 diabetes mellitus
Repetitive episodes of moderate to severe hypoglycemia
Memory* have been implicated as one possible etiology of cognitive
Verbal memory dysfunction in diabetes. This is significant because the risk of
Visual retention hypoglycemia increases as efforts to achieve the level of
Working memory
Immediate recall
glycemia necessary to minimize the risk of developing
Delayed recall the microvascular complications of diabetes are intensified
Psychomotor speed* (56 –58). The reason for severe hypoglycemia secondary to
Executive function* intensive insulin management is complex and multifactorial,
Processing speed however the initial intelligence of patients with type 1 dia-
Complex motor function
Verbal fluency betes before intensive management does not predispose to
Attention more future hypoglycemia episodes, as shown by an analysis
Depression of data collected during the DCCT (59). During acute hypo-
Domains marked by asterisks have particularly strong support- glycemia episodes, it has been shown that performance
ing data. on immediate verbal memory, immediate visual memory,

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 22 May 2014. at 00:09 For personal use only. No other uses without permission. . All rights reserved.
Kodl and Seaquist • Cognition and Diabetes Mellitus Endocrine Reviews, June 2008, 29(4):494 –511 497

working memory, delayed memory, visual-motor skills, visual- frequent episodes of hypoglycemia (⬍70 mg/dl) actually
spatial skills, and global cognitive dysfunction are all impaired had increased memory and verbal scores, as well as overall
(60, 61). In a more recent study, prospective memory (that is, better academic achievement when compared with less well-
“remembering to remember”) and immediate and delayed re- controlled children with diabetes.
call were both impaired secondary to hypoglycemia in patients
with type 1 diabetes, suggesting that impairments with both D. Section summary
recall and learning/consolidation occur during hypoglycemia
(62). Interestingly, in some studies there was no difference in Clearly, much research has been done on cognitive dys-
reaction time (63), memory (62), and overall cognitive perfor- function in patients with type 1 and type 2 diabetes mellitus.
mance (61) between hypoglycemia aware and unaware patients Although results are not consistent and many different def-
during hypoglycemic episodes, despite the fact that the glucose icits have been identified, some conclusions can be drawn. In
level at which the counterregulatory hormone response was patients with type 1 diabetes mellitus, deficits in speed of
elicited was higher in subjects with awareness of hypoglycemia. information processing, psychomotor efficiency, attention,
Although cognitive impairment may occur during hypo- mental flexibility, and visual perception seem to be present,
glycemia, the effect of repetitive hypoglycemia on subse- whereas in patients with type 2 diabetes, an increase in
quent cognitive function during euglycemia is less clear. memory deficits, a reduction in psychomotor speed, and
Studies have shown impaired verbal IQ scores (14, 64), full reduced frontal lobe/executive function have been identi-
scale IQ scores (14, 20, 64), attention (20), verbal skills (11), fied. Severe hypoglycemic episodes may contribute to cog-
short-term memory, verbal memory (17), vigilance (65), and nitive dysfunction in the young; however, as patients age
visual-spatial memory (8, 18) in patients with a history of episodes seem to have less of an influence. Finally, improved
type 1 diabetes and severe hypoglycemia (defined as being diabetes control and decreased diabetic complications seem
associated with seizures, coma, or the need for external as- to be associated with less cognitive dysfunction, although
sistance), compared with patients with type 1 diabetes with- this association is clearer in patients with type 2 diabetes than
out a history of severe hypoglycemia. However, it is possible with type 1 diabetes.
that some of these “abnormalities” could be the result of However, some questions remained unanswered. First, it
slower, more deliberate completion of the tasks without loss is not clear whether cognitive impairments seen in neuro-
of accuracy (66). More recently, no association between mul- cognitive testing result in meaningful deficits either socially
tiple severe episodes of hypoglycemia and impaired cogni- or professionally. Given the subjective nature of assessing
tive function in patients with type 1 diabetes mellitus was professional and social activities, it will be difficult to address
found in an 18-yr follow-up of the DCCT (15). Although the this question. Second, although the data suggest that hyper-
DCCT follow-up has been regarded as a landmark study that glycemia contributes to cognitive impairment, the magni-
provides reassurance to diabetologists and patients, it is im- tude of this contribution and how hyperglycemic one must
portant to recognize its limitations, including the fact that it be to experience the ill effects of hyperglycemia on cognition
did not randomize patients into a “severe hypoglycemia” are not clear. Lastly, it is unknown whether mild neurocog-
group and a control group (because such randomization nitive impairments will progress to overt dementia. Large
would be not only logistically impossible but also unethical). randomized controlled trials such as the Epidemiology of
A lack of association between severe hypoglycemia and cog- Diabetes Interventions and Complications (EDIC) study/
nitive dysfunction was confirmed by other studies (23, 68 – DCCT and the ongoing Action to Control Cardiovascular
71), as well as with a meta-analysis, which showed no as- Risk in Diabetes (ACCORD) study should hopefully con-
sociation between hypoglycemia and cognitive function (22). tinue to address these last two questions.
Of note, however, most data analyzing the effects of hypo-
glycemia look at young to middle-aged patients; data re-
III. Pathophysiology of Cognitive Dysfunction in
garding the impact of hypoglycemia on older individuals is
Diabetes
lacking.
One possible reason that some studies found an associa- The pathophysiology underlying the development of cog-
tion between frequent hypoglycemia and cognitive dysfunc- nitive dysfunction in patients with diabetes has not been
tion and others did not is that the positive investigations may completely elucidated. Many hypotheses with supporting
have included subjects with diabetes onset earlier in life. evidence exist, including potential causative roles for hyper-
Patients with type 1 diabetes diagnosed at less than 5 yr of glycemia, vascular disease, hypoglycemia, insulin resistance,
age may have more severe (often with seizures) and frequent and amyloid deposition (Fig. 1). Although further research
hypoglycemia episodes than those diagnosed at ages older into each of these candidate mechanisms is necessary, it may
than 5 yr; these younger patients have been found to have be that the cause of cognitive dysfunction in patients with
worse cognitive dysfunction (9, 18, 72, 73). The severity of the diabetes will turn out to be a combination of these factors,
hypoglycemia as well as the susceptibility of young brains to depending on the patient’s type of diabetes, comorbidities,
injury may explain the discrepancy (9, 74). Another expla- age, and type of therapy.
nation for discrepancy between reports is that subjects with
more hypoglycemia may have overall tighter glycemic con- A. The role of hyperglycemia
trol, which may offset the neurocognitive damage from hy-
poglycemia. This was most likely the case in the population As reviewed in Sections II.A and II.B, hyperglycemia ap-
studied by Kaufman et al. (17) in which children with more pears to be related to abnormalities in cognitive function in

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 22 May 2014. at 00:09 For personal use only. No other uses without permission. . All rights reserved.
498 Endocrine Reviews, June 2008, 29(4):494 –511 Kodl and Seaquist • Cognition and Diabetes Mellitus

Hyperglycemia-
Induced End Organ
Damage
“Microvascular Disease”

“Macrovascular
Insulin Resistance
Disease”
Cerebrovascular
Accident

Cognitive Dysfunction in
Diabetes Mellitus

Absence of
Hypoglycemia
C-Peptide

Absence of
Apoε 4 Allele

FIG. 1. Summary of possible mechanistic contributors to cognitive dysfunction seen in diabetes mellitus. Not all mechanisms are present in
every patient.

patients with both type 1 and type 2 diabetes. However, the function normally seen in rats with hyperglycemia (80).
mechanisms through which hyperglycemia might mediate Whether this pathway contributes to neurocognitive dys-
this effect are less than clear. In other organs, hyperglycemia function in humans with diabetes is unknown.
alters function through a variety of mechanisms including The role of AGEs and receptors for AGE (RAGEs) in the
polyol pathway activation, increased formation of advanced development of cerebral complications of diabetes also re-
glycation end products (AGEs), diacylglycerol activation of mains uncertain. Diabetic mice (32% HbA1c vs. 12% in con-
protein kinase C, and increased glucose shunting in the hex- trol mice) with demonstrated cognitive impairment have
osamine pathway (75–78). These same mechanisms may be been found to have increased expression of RAGEs in neu-
operative in the brain and induce the changes in cognitive rons and glial cells and damage to white matter and myelin
function that have been detected in patients with diabetes. (78), suggesting a possible role of RAGEs in the development
It has long been known that hyperglycemia increases of cerebral dysfunction (81). In humans, patients with dia-
flux through the polyol pathway in nervous tissue. In the betes and Alzheimer’s disease have been found to have
streptozotocin-treated rat (glucose concentration 27.4 ⫾ 0.3 greater N-carboxymethyllysine (a type of AGE) staining on
mmol/liter vs. 5.9 ⫾ 0.1 mmol/liter in control rats), an in- brain slices obtained postmortem than patients with Alzhei-
crease in sorbitol was measured in cranial nerves, sciatic mer’s disease alone (82). However, a second autopsy study
nerve, cerebral cortex, and retina. This accumulation was failed to find a difference in the quantity of AGE-like glycated
reduced significantly when the animals were treated with the protein rich neurofibrillary tangles and senile plaques, like
aldose reductase inhibitor tolerstat (79). Another study look- those seen in patients with Alzheimer’s disease, between
ing at streptozotocin-treated rats (HbA1c 7.9 ⫾ 0.3 vs. 3.3 ⫾ human subjects with diabetes and controls (83). Experiments
0.0 in control rats) also found that administering an aldose performed in animal models provide limited evidence to
reductase inhibitor, sorbinil, reduced the accumulation of support the hypothesis that AGE-induced brain injury may
brain tissue sorbitol and corrected the reduced cognitive be a mechanism through which hyperglycemia and diabetes

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 22 May 2014. at 00:09 For personal use only. No other uses without permission. . All rights reserved.
Kodl and Seaquist • Cognition and Diabetes Mellitus Endocrine Reviews, June 2008, 29(4):494 –511 499

alter cerebral function. In vitro the addition of AGEs to bovine acetylcholine (91), decreased serotonin turnover, decreased
brain microvascular endothelial cells up-regulates both tis- dopamine activity, and increased norepinephrine (86, 92) in
sue factor mRNA (which induces blood coagulation) and the brains of animals with diabetes. Interestingly, these changes
reactive oxygen species through a mechanism that is re- were all reversed with insulin. One proposed hypothesis is that
versed with treatment by the free radical scavenger edara- the alternating high and low glucose levels seen in patients
vone (84). In addition, in a rat model of focal cerebral isch- with poorly controlled diabetes may worsen neurotransmitter
emia, the infusion of AGEs increased cerebral infarct size, function (92).
whereas the coadministration of aminoguanidine, an inhib-
itor of AGE cross-linking, attenuated the infarct volume (85).
Few investigators have examined the role of diacylglycerol B. The role of vascular disease
activation of protein kinase C and increased glucose shunting
in the development of cognitive dysfunction in diabetes. Patients with diabetes have a 2- to 6-fold increased risk in
Brain expression of protein kinase C-␣ was shown to be thrombotic stroke (41, 93), and vascular disease has long been
significantly increased in one untreated diabetic rat model hypothesized to contribute to abnormalities in cognition in
(approximate blood glucose, 15 mmol/liter) compared with such patients. Autopsy studies on patients with long-stand-
the treated diabetic rat and control rats (approximate blood ing type 1 diabetes have shown changes related to vascular
glucose, 6 mmol/liter) (86), but another study found no disease, including diffuse brain degeneration, pseudocalci-
increase in protein kinase C activity in diabetic rats (approx- nosis, demyelination of cranial nerves and spinal cord, and
imate blood glucose, 17 mmol/liter) compared with controls nerve fibrosis (94, 95). Thickening of capillary basement
(approximate blood glucose, 6.4 mmol/liter) (87). Support membranes, the hallmark of diabetic microangiopathy, has
for the possible role of the hexosamine pathway comes from
been found in the brains of patients with diabetes (96). Pa-
the interesting observation that cerebral chitin, an N-acetyl-
tients with diabetes have also been found to have decreased
glucosamine polymer produced via the hexosamine path-
global rates of cerebral blood flow as measured using xenon,
way, is increased in human subjects with Alzheimer’s disease
on autopsy (88). If hyperglycemia from diabetes shunts glu- and the magnitude of reduction correlates with the duration
cose toward the production of chitin, it is possible that the of the disease. However, blood glucose levels were not con-
accumulation of this molecule could contribute to abnormal- trolled during the experiment (range, 3.1–21.2; mean, 8.8 ⫾
ities in cognition. 4.74 mmol/liter) (97). Interestingly, the rate of cerebral blood
Hyperglycemia has also been proposed to cause end organ flow in patients with diabetes is similar to that found in
damage through increases in reactive oxygen species, in par- Alzheimer’s patients with dementia (92). These observations
ticular superoxide, which could then lead to increased polyol in humans with diabetes are supported by studies in strep-
pathway activation, increased formation of AGEs, activation tozotocin-treated rats with chronic hyperglycemia (mean
of protein kinase C, and increased glucose shunting in the plasma glucose, approximately 29 mmol/liter) (98). One can
hexosamine pathway (76). Using streptozotocin to induce speculate that the decrease in cerebral blood flow, coupled
diabetes in rats (blood glucose, 20.72 ⫾ 2.25 vs. 6.04 ⫾ 0.64 with the stimulation of the thromboxane A2 receptor known
mmol/liter in control rats), Aragno et al. (89) found that to occur in patients with diabetes (92), could contribute to the
RAGEs, galectin-3 (a proatherogenic molecule), and the inability of cerebral vessels to adequately vasodilate, which
polyol pathway activation all were increased in rat brains, may in turn increase the likelihood of ischemia. The coex-
whereas activity of the glycolytic enzyme glyceradehyde-3- istence of ischemia and hyperglycemia may be particularly
phosphate dehydrogenase was decreased, indicating ele- detrimental to the brain. Even modestly elevated blood glu-
vated superoxide levels. Nuclear factor ␬B transcription fac- cose levels (greater than 8.6 mmol/liter) in humans during
tors, a proinflammatory gene marker up-regulated by AGEs, a cerebrovascular event correlates with poorer clinical re-
and S-100 protein, a marker for brain injury that can bind to covery (99). One potential mechanism through which hy-
RAGEs, were both up-regulated in the hippocampus in this perglycemia could potentiate ischemic damage is lactate ac-
animal model, although the effect in other regions was not cumulation. Hyperglycemia provides more substrate for
assessed. These data suggest that oxidative stress may trigger
lactate to form, causing cellular acidosis and worsening in-
a cascade of events that lead to neuronal damage. Interest-
jury (93). Another mechanism is the accumulation of gluta-
ingly, dehydroepiandrosterone, an adrenal androgen and
mate in the setting of hyperglycemia and ischemia (100).
antioxidant, significantly reduces these changes, suggesting
a potential therapy worthy of more investigation. Glutamate, an excitatory amino acid neurotransmitter, has
In addition to hyperglycemic-induced end organ damage, been shown to cause neuronal damage in the brain (101).
altered neurotransmitter function has been observed in di- Although the exact mechanism is not known, the lack of
abetic models and may also contribute to cognitive dysfunc- C-peptide in patients with type 1 diabetes may by itself
tion. In diabetic rats (blood glucose 28.6 ⫾ 1.1 vs. 6.3 ⫾ 0.2 worsen cognitive impairment through its actions on the en-
mmol/liter in control rats), there is an impairment of long- dothelium. Evidence for this is suggested by a rat model
term potentiation, defined as activity-dependent prolonged (blood glucose approximately 23 mmol/liter) in which re-
enhancement of synaptic strength, in neurons rich in recep- placement of C-peptide to normal levels normalized cogni-
tors for the neurotransmitter N-methyl-d-aspartate (NMDA), tive function and reduced hippocampal apoptosis (102). The
which could contribute to learning deficits (90). Other, neuro- relevance to humans is uncertain, however, because humans
chemical changes have been observed, including decreased with type 1 diabetes do not have hippocampal atrophy (103).

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 22 May 2014. at 00:09 For personal use only. No other uses without permission. . All rights reserved.
500 Endocrine Reviews, June 2008, 29(4):494 –511 Kodl and Seaquist • Cognition and Diabetes Mellitus

C. The role of hypoglycemia found to be widely distributed in rat brain using immuno-
histochemistry and in situ hybridization (114, 115), respec-
As mentioned in Section II.C, whether repeated episodes of
tively, including in the olfactory bulb, hypothalamus, hip-
hypoglycemia contribute to cognitive dysfunction is contro-
pocampus, cerebellum, piriform cortex, cerebral cortex, and
versial and most likely depends on the age of the patient.
amygdala. The insulin-responsive glucose transporter 4
However, there is no argument that if severe hypoglycemia
(GLUT4) has also been found in select regions of the rat brain,
lasts for a very long time, brain damage and death can occur
including the pituitary, hypothalamus, and medulla (116).
(93, 104 –106). Most endocrinologists have personal experi-
GLUT8, also known as GLUTx1, is also found in the rat brain,
ence with patients who have experienced severe hypogly-
specifically in the hippocampus, hypothalamus, cerebellum,
cemia (⬍2 mm) with little or no permanent consequence. This
and brainstem (117). GLUT8 has similar properties to GLUT4
most likely is secondary to inaccuracies of glucometer at low
and is up-regulated in response to insulin in some (118) but
blood glucose levels (107), inadequate time with severe hy-
not all murine tissues, including the brain (119). Despite the
poglycemia, or variations in patients’ glycogen stores. That
presence of insulin receptors and insulin-sensitive glucose
said, it has been shown in animal models that after 30 – 60 min
transporters, the effect of insulin on cerebral glucose metab-
of blood glucose levels between 0.12 and 1.36 mmol/liter,
olism is still uncertain. Many laboratories, including our
neuronal necrosis occurs with accompanying extracellular
own, have failed to demonstrate an effect of insulin on ce-
increases in aspartate, alkalemia, and neuronal energy fail-
rebral glucose metabolism in humans (120 –122). However,
ure, ultimately leading to a flat electroencephalograph (EEG)
other laboratories using fluorodeoxyglucose positron emis-
(105, 106). The cortex, basal ganglia, and hippocampus ap-
sion tomography (PET) have found a significant increase in
pear to be most vulnerable to hypoglycemia, with laminar
brain glucose metabolism in the setting of hyperinsulinemia
necrosis and gliosis found in these regions on autopsies per-
in humans (123), an effect that is reduced in subjects with
formed in human patients who died of hypoglycemia (104).
peripheral insulin resistance (124). Despite the ongoing con-
Other human autopsy studies done after death secondary to
troversy of the effect of insulin on cerebral glucose metab-
hypoglycemia have shown multifocal or diffuse necrosis of
olism, there is a large and growing body of evidence that
the cerebral cortex and chromatolysis of ganglion cells (108).
insulin resistance, long recognized as a factor contributing to
In animal models, hypoglycemia-induced damage seems to
the onset of type 2 diabetes, may play a role in the patho-
be selective to neurons with sparing of astrocytes and oli-
genesis of Alzheimer’s disease.
godendrocytes (92). Although counterintuitive, the time to
The clinical diagnosis of Alzheimer’s disease is made in the
neuronal death may be asymmetric between hemispheres in
presence of a significant gradual and progressive decline in
severe, prolonged hypoglycemia, making the differentiation
memory with at least one other cognitive, social, or occupa-
of hypoglycemic brain damage from ischemia difficult on a
tional disturbance (125). The incidence of Alzheimer’s dis-
clinical basis (105). Some have hypothesized that hypogly-
ease has been found to be approximately 1.2- to 1.7-fold
cemia-induced neuronal damage occurs as a result of over-
higher in patients with type 2 diabetes and insulin resistance
activation of a subtype of the excitatory neurotransmitter
compared with a control population in most (37– 43, 47), but
NMDA receptor (109). Interestingly, there exists an NMDA
not all (126 –128), investigations. The reason for the discrep-
receptor antagonist that has been shown to prevent neuronal
ancy could be the populations studied. Data that support a
necrosis, suggesting a potential therapy for hypoglycemia-
relationship between Alzheimer’s and insulin resistance all
induced brain damage (110). Such a therapy may be helpful
examined older subjects ascertained from the general pop-
in young children with type 1 diabetes who seem to be
ulation. The reports that failed to find an association collected
particularly susceptible to cerebral complications of hypo-
data from a more narrowly defined population, such as
glycemia. There may also be a relationship to hypoglycemia
those with a high incidence of the apolipoprotein E type 4
during early nocturnal sleep, a time in which consolidation
(APOE-␧4) allele (128) or a high percentage of early-onset
of memories occurs, and cognitive dysfunction. Compared
Alzheimer’s disease (126). Interestingly, it appears that
with test outcomes after a night of sleep in euglycemia,
type 2 diabetes is also more common in populations with
human control subjects and subjects with type 1 diabetes
Alzheimer’s disease (129). Whether the association between
exhibited impaired declarative memory (memory of facts)
Alzheimer’s disease and patients with type 2 diabetes reflects
after undergoing a short, relatively mild hypoglycemic
the impact of poor metabolic control on brain function or the
clamp (2.2 mmol/liter) during early sleep (111). However, no
actual effects of insulin and insulin resistance on the brain is
neurocognitive deficits were seen in several other studies in
unclear. Patients with Alzheimer’s disease and normal glu-
which nocturnal hypoglycemia was induced later during the
cose tolerance have a more robust insulin secretory response
sleeping period (112, 113).
to an oral glucose load than controls, suggesting that they
may have increased insulin resistance (130, 131). Some have
D. The role of insulin resistance and amyloid suggested that the insulin resistance occurs in the brain itself
and have hypothesized that the desensitization of neuronal
Although the role of insulin on cerebral metabolism and insulin receptors plays an important role in the development
function is still evolving, fascinating research has given us of sporadic Alzheimer’s disease (132). Such a concept is sup-
more insight into this field over the last 20 yr. Historically, ported by observations that patients with Alzheimer’s dis-
the brain was thought to be an insulin-independent organ; ease have an elevated concentration of insulin in their cere-
however, many recent discoveries have questioned that no- bral spinal fluid under fasting conditions (131), along with an
tion. Insulin receptors and mRNA expression have been increase in insulin receptor density in the occipital region and

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 22 May 2014. at 00:09 For personal use only. No other uses without permission. . All rights reserved.
Kodl and Seaquist • Cognition and Diabetes Mellitus Endocrine Reviews, June 2008, 29(4):494 –511 501

a decrease in tyrosine kinase activity (which is downstream ment for 8 wk improved delayed recall, enhanced mood, and
to insulin binding) in the temporal and occipital lobes com- self confidence and reduced anger in nondiabetic, nonde-
pared with controls (133). However, others have found that mentia subjects (151). Based on these studies, it is hypothe-
patients with Alzheimer’s disease have a decrease in cerebral sized that in Alzheimer’s disease, cerebral insulin resis-
spinal fluid insulin levels, suggesting that there may be im- tance requires higher levels of insulin to facilitate memory
paired insulin transport across the blood brain barrier or (46, 152). Although cerebral insulin is higher in these pa-
increased insulin catabolism that accounts for the impaired tients, it may not be enough to compensate for the insulin
central insulin action (134). resistance. However, this does not necessarily prove that
The mechanisms through which insulin resistance might hyperinsulinemia directly improves cognitive function. Hy-
alter cognitive function remain uncertain, but effects on neu- perinsulinemia can stimulate epinephrine release, and both
rotransmission and memory formation have been hypothe- insulin and epinephrine have been shown to increase lactate
sized. An impairment in central cholinergic activity is be- (153). Lactate can then in turn be used as a source of energy
lieved to contribute to the pathogenesis of Alzheimer’s in brain metabolism (154, 155), although the benefits of lac-
disease (135), and interestingly, rats with streptozotocin- tate therapy have not been proven to be beneficial yet in
induced diabetes have a decrease in the production and Alzheimer’s patients (156).
release of acetylcholine compared with control rats (91). Mice Insulin resistance and type 2 diabetes mellitus may con-
models in which cholinergic activity is blocked by scopol- tribute to cognitive dysfunction through three other indirect
amine experience amnesia and hyperactivity, a deficit that mechanisms. First, cognitive dysfunction in patients with
can be reversed by glucose administration (136, 137). Glucose type 2 diabetes has been correlated to inflammatory markers,
administration with a rise in endogenous insulin levels or and increased inflammation may contribute to the develop-
insulin administration to patients with Alzheimer’s disease ment of Alzheimer’s or macrovascular disease. In one in-
have also been shown to alter behavior, perhaps by enhanc- vestigation, patients with the metabolic syndrome, elevated
ing cholinergic activity (138). In these studies, patients with C-reactive protein, and elevated IL-6 were found to have
Alzheimer’s demonstrated an improvement in declarative impaired cognitive function, whereas those patients with
memory during either a hyperglycemic or a hyperinsuline- the metabolic syndrome and normal levels of these inflam-
mic euglycemic clamp (138, 139). Furthermore, patients with
matory markers had similar cognition to controls (47). Pa-
memory impairment and Alzheimer’s disease had improved
tients with type 2 diabetes are known to have higher levels
verbal memory acutely after intranasal insulin administra-
of inflammatory markers including C-reactive protein, ␣-
tion, which had no effect on peripheral glucose or insulin
1-antichymotrypsin, IL-6, and intercellular adhesion mole-
levels but had previously been shown to increase central
cule 1 than control populations (157). These findings raise the
nervous system insulin levels (140). In addition to affecting
possibility that insulin resistance and Alzheimer’s disease
cholinergic activity, diabetes and insulin may affect long-
share a common pathophysiology, because patients with
term potentiation in opposing ways. Long-term potentiation
Alzheimer’s disease demonstrate increased inflammatory
is critical to the formation of memories and is induced by
NMDA receptor activation, a process that is up-regulated in markers as well (158, 159).
the presence of insulin (141). However, rats with diabetes, A second potential mechanism through which insulin re-
and presumed relative insulin deficiency, have decreased sistance and type 2 diabetes could contribute to cognitive
long-term potentiations in the hippocampus as measured by dysfunction is through the disruption of the hypothalamic-
electrophysiology (90). As would be expected if long-term pituitary adrenal axis. Both animals (160) and humans (161)
potentiation were reduced, rat hippocampal neurons ex- with type 2 diabetes have an up-regulation of the hypotha-
posed to insulin exhibited inhibition of spontaneous firing lamic-pituitary-adrenal axis, with increased serum cortisol
(142). Interestingly, patients with Alzheimer’s disease have compared with controls. In other research, hypercortisolemia
a reduced cerebral glucose uptake as measured by PET has been found to cause cognitive dysfunction. Healthy hu-
(143–145) and have a reduced number of glucose transport- mans treated with dexamethasone (162), corticosterone (163),
ers in the brain microvessels, frontal cortex, hippocampus, and hydrocortisone (164) to mimic stress conditions all per-
caudate nucleus, parietal, occipital, and temporal lobe com- formed worse on memory testing. In a study of healthy
pared with controls on autopsy studies (146, 147). Perhaps elderly patients, those with higher serum cortisol levels per-
the reduction in glucose uptake has a direct effect on how formed more poorly on memory and attention testing (165).
insulin regulates hippocampal function in these patients. In addition, patients with Cushing’s disease have been found
Future experiments to identify the relative roles of glucose to have worse performance on memory, attention, reasoning,
and insulin in human cognition are necessary to clarify these and concept formation testing compared with controls (166),
relationships. which may be attributed to a significant reduction in cerebral
The impact of insulin on cognitive function has also been glucose metabolism found on PET scan in those patients with
examined in control subjects without Alzheimer’s disease or Cushing’s disease (167). Supporting these findings are the
diabetes mellitus. In our laboratory, we found that inducing animal studies in which glucocorticoids cause structural
hyperinsulinemia using an insulin infusion in control sub- damage and reduce function of neurons in the hippocampus
jects reduces parietal region P300 amplitude secondary to (168 –172). Based on the facts that type 2 diabetes causes an
memory triggers (148). Other clamp studies found improved up-regulation of the hypothalamic-pituitary-adrenal axis
vigilance, memory, and selective attention in the setting of and hypercortisolemia can cause cognitive dysfunction, it
hyperinsulinemia (149, 150), whereas intranasal insulin treat- can be hypothesized that the increase in cortisol levels seen

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 22 May 2014. at 00:09 For personal use only. No other uses without permission. . All rights reserved.
502 Endocrine Reviews, June 2008, 29(4):494 –511 Kodl and Seaquist • Cognition and Diabetes Mellitus

in patients with type 2 diabetes may contribute to cognitive shared pathogenesis may be present in patients with type 2
dysfunction. diabetes and patients with Alzheimer’s disease, possibly in-
The third potential mechanism through which insulin re- volving a defect in a chaperone protein (191) that helps
sistance may indirectly contribute to cognitive dysfunction intracellular protein trafficking (129). Rat models of type 2
is by promoting the formation of senile plaques found in diabetes (BBZDR/Wor), even more so than type 1 (BB/Wor)
Alzheimer’s disease. Intracellular neurofibrillary tangles and diabetes, demonstrate an increase in Alzheimer’s pathology,
extracellular senile plaques composed of ␤-amyloid are the including increased APP, ␤-amyloid, and ␤-secretase and
pathological hallmarks of Alzheimer’s disease (173–175). loss of neurons (192). Despite this compelling evidence link-
␤-Amyloid is formed from the cleavage of amyloid precursor ing insulin resistance and type 2 diabetes mellitus to Alz-
protein (APP), produced in neurons (176), by the enzymes heimer’s disease and pathology, several autopsy studies per-
␤- and ␥-secretase (177). ␤-Amyloid is eventually degraded formed in humans have failed to identify an increase in senile
by the insulin-degrading enzyme (178, 179). Amyloid ␤- plaques or neurofibrillary tangles in subjects with diabetes
peptides can by themselves bind to RAGEs and bring about compared with age-matched controls (83, 129), although the
microglial and neuronal dysfunction and oxidative stress duration of diabetes did correlate with the density of senile
(81). Interestingly, amyloid ␤-peptides, AGEs, and RAGEs plaques in one of these studies (129).
have all been colocalized in astrocytes using immunohisto- The relationship between insulin resistance and cognitive
chemistry in human brain slices (180). In addition, there is a dysfunction in Alzheimer’s disease appears to depend on the
growing body of evidence that insulin and insulin resistance presence or absence of the APOE-␧4 allele. Curiously, al-
can affect the metabolism of APP and ␤-amyloid, thus po- though the presence of the APOE-␧4 allele is associated with
tentially increasing the burden of cerebral senile plaques. The an increased incidence of Alzheimer’s disease (193), it seems
role of insulin resistance in the metabolism of APP and that insulin resistance is only a significant risk factor for
␤-amyloid was further clarified by Craft et al. (181). In their Alzheimer’s disease in those patients without the APOE-␧4
experiment, plasma levels of APP, the precursor to ␤-amy- allele (39, 134). Patients with Alzheimer’s disease and no
loid, were lower in those subjects with insulin resistance and APOE-␧4 allele have been observed to have lower glucose
Alzheimer’s disease when undergoing a hyperinsulinemic- disposal rates during a hyperinsulinemic-euglycemic clamp
euglycemic clamp. This corresponded with improved mem- than subjects with Alzheimer’s disease and the APOE-␧4
ory testing. One potential explanation of this observation is allele, as well as those subjects without Alzheimer’s disease
that insulin resistance may cause decreased APP degradation or the APOE-␧4 allele. Subjects with Alzheimer’s disease
that can be overcome by the elevating serum and presumably without the APOE-␧4 allele also had improved memory
tissue insulin levels (181). Similar findings have come from scores in the setting of hyperinsulinemia, which was not the
experiments in rat hippocampal neurons, where insulin was case in APOE-␧4 allele-positive subjects (149, 181). Based on
found to up-regulate insulin degrading enzyme, thereby in- this information, it seems that insulin resistance/type 2 di-
creasing ␤-amyloid degradation (182). However, not all stud- abetes and APOE-␧4 allele positivity are distinct and separate
ies have agreed with this hypothesis. In a study using neu- risk factors for the development of Alzheimer’s disease, a
roblastoma cell lines by Gasparini et al. (178), insulin was hypothesis that is supported by the fact that those with
found to decrease intracellular ␤-amyloid and increase ex- diabetes had a low incidence of the APOE-␧4 allele (128).
tracellular levels of ␤-amyloid by both promoting its secre- However, there is again a contradiction in the literature; in
tion and inhibiting its degradation via the insulin-degrading the Honolulu-Asia Aging Study, those subjects with both
enzyme. This would contradict the majority of the evidence type 2 diabetes and the APOE-␧4 allele had an additive
that insulin has a protective effect against memory loss. increased risk of dementia and Alzheimer’s pathology (43).
However, whereas it is widely believed that extracellular This study was specific for elderly Japanese-American men,
accumulation of ␤-amyloid plays a critical role in the devel- so it seems that additional multiethnic studies are needed to
opment of Alzheimer’s, other evidence is suggesting there is understand this discrepancy better.
a pathogenic role for intracellular ␤-amyloid (183–185). More The association between insulin resistance and Alzheimer’s
research is needed concerning the pathophysiology of disease has been sufficiently compelling for investigators
␤-amyloid and insulin before conclusions can be drawn. to examine whether peroxisome proliferator-activated
Of interest is the observation that the pancreatic islets in receptor-␥ (PPAR-␥) agonists can treat Alzheimer’s disease
patients with type 2 diabetes mellitus are characterized by in the absence of diabetes. To date, two trials have demon-
␤-cell loss and deposition of islet amyloid (186), which is strated rosiglitazone to have a beneficial effect on memory in
reminiscent of the neuronal loss and ␤-amyloid deposition patients with Alzheimer’s disease. In a small randomized
seen in Alzheimer’s disease (187). The constitutions of islet study published by Watson et al. (194) in 2005, patients with
and neural ␤-amyloid are similar (129, 188), and both are mild Alzheimer’s disease treated for 6 months with rosigli-
toxic to islet and neurons, respectively (189, 190). In a series tazone had better memory and selective attention than con-
of 29 patients in whom both brain and pancreas autopsy trols. A much larger study published in 2006 found that
specimens were available, all had amyloid detected to some patients with Alzheimer’s disease without the APOE-␧4 al-
degree in both the brain and pancreas (187). In another study, lele had improvements in cognitive testing after 6 months of
islet amyloid was more abundantly present on autopsy in rosiglitazone, whereas those Alzheimer’s disease patients
patients with Alzheimer’s disease than in those without with the APOE-␧4 allele did not have improvements (195).
Alzheimer’s disease (129). Based on the similarity between Multiple mechanisms have been proposed to address how
islet and neural ␤-amyloid, some have speculated that a PPAR-␥ agonists may affect the pathophysiology responsible

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 22 May 2014. at 00:09 For personal use only. No other uses without permission. . All rights reserved.
Kodl and Seaquist • Cognition and Diabetes Mellitus Endocrine Reviews, June 2008, 29(4):494 –511 503

for Alzheimer’s disease, including reducing serum glucocorti- TABLE 3. Summary of modalities for assessment of cognitive
coids (196), decreasing glial inflammation (197, 198), protecting dysfunction in patients with diabetes
against ␤-amyloid-induced neurodegeneration (199), decreas-
Neurocognitive testing
ing ␤-amyloid production (197), increasing ␤-amyloid degra- Evoked response potentials
dation (196, 200), and decreasing phosphorylation of tau pro- EEG
teins, the mechanism by which neurofibrillary tangles are MRI
formed (201). Interestingly, despite all of these data, it is still fMRI
SPECT
not totally clear how rosiglitazone benefited patients with PET
Alzheimer’s disease in clinical trials because it has been shown
not to cross the blood-brain barrier (196). These data are com-
pelling enough to warrant further, longer-term studies on the tentials can reveal subclinical sensory nerve conduction def-
benefits of PPAR-␥ agonists in the treatment of Alzheimer’s icits that may not otherwise be apparent (206). For example,
disease. However, clinicians must weight the benefits against flash electroretinography has shown decreased potentials
the newly documented cardiovascular risks of these treatments from the retina in diabetic subjects before ophthalmoscopic
(202, 203). signs of retinopathy were seen (108). In addition, pattern
electroretinogram, which looks at the pattern of retinal stim-
uli originating from the ganglion cells, is also decreased in
IV. Modalities for Assessment of Cognitive patients with diabetes (108). The evoked response of nerves
Dysfunction in Patients with Diabetes involved in sensing auditory stimuli is also abnormal; brain-
stem auditory-evoked potentials demonstrated acoustic
Although progress is being made, the difficulty of detect- pathway impairment in patients with diabetes (207–209).
ing neurocognitive dysfunction in patients with diabetes in Evoked potentials related to memory may also be affected
the clinical setting may explain in part why the field of because auditory P300 event-related potentials had signifi-
cognitive dysfunction in diabetes has not advanced similarly cantly longer latencies in patients with type 2 diabetes com-
to other fields dealing with hyperglycemia-associated end pared with controls, which could relate to attention and
organ damage. Neurocognitive testing in which an examiner short-term memory defects (210). Central somatosensory-
administers a battery of tests to assess different aspects of evoked potentials were found to be prolonged in patients
cerebral function has long been the gold standard for the with diabetes as well (211). In another study looking at both
assessment of neurocognitive function. Although cumber- type 1 and type 2 diabetes, slowed latency in visual and
some to administer and score, it has been very useful in somatosensory-evoked potentials was observed in patients
assessing neurocognition in a variety of disease states, in- with type 1 diabetes, whereas patients with type 2 diabetes
cluding diabetes, as was demonstrated in Section II. How- had slowed latency of visual, somatosensory, and brainstem
ever, such tests have a relatively high rate of intrasubject auditory-evoked potentials (212). In this investigation, in-
variability that reduces their ability to identify mild deficits creasing HbA1c was related to reduced cognitive perfor-
or preclinical disease. Also, many studies examining the mance. Event-related potentials have also helped define
effect of diabetes on brain function use multiple neurocog- brain adaptations to hypoglycemia. During hypoglycemia,
nitive tests that assess the same psychological process. When normal subjects do not experience a delay in initial percep-
the results of these different tests don’t agree, determining tion and precognitive processing, but they do have a delay
which results to base conclusions on can be confusing (204). in central processes such as stimulus selection and selective
In addition, not all neurocognitive tests are created equal. central motor activation (213). Of note, although all except
Although many neurocognitive tests are well validated in a one of these studies controlled for hypoglycemia during
diverse population to distinguish between “normal” and testing (211), none of the studies adequately controlled for
“abnormal” results, other tests do not have adequate reli- hyperglycemia during testing, although Kurita et al. (210)
ability data, are based on unacceptably small norms, are found no difference between those subjects with high and
administered inappropriately, or do not properly distinguish relatively normal blood glucose values.
between two or more diagnostic groups (205). Finally, neu- EEG can also assess spontaneous cerebral electrical activ-
rocognitive testing is unable to provide specific information ity and has been used in patients with type 1 and type 2
about the neural structures responsible for any dysfunction diabetes. Patients with type 2 diabetes have been found to
identified. For example, although it appears that white mat- have slowing in the EEG frequency band analysis over the
ter function such as processing speed, attention, and visual- central cortex area and reduction of alpha activity over the
spatial processing are particularly affected by diabetes (4), parietal area. These findings correlated with reduced visual
localization of this dysfunction to white or gray matter is not retention on neurocognitive testing but were not simply re-
possible using the battery of tests available to assess lated to hyperglycemia because making nondiabetic subjects
neurocognition. with hyperglycemia did not reproduce these findings (32).
Because of the limitations in neurocognitive testing, a Subjects with type 1 diabetes have also been found to have
number of modalities have been used to assess cognitive abnormal EEG results compared with controls, with those
function in patients with diabetes (Table 3). One of the oldest patients with a history of having severe hypoglycemia hav-
modalities has been to measure electrical activity such as ing the most abnormalities (65, 214, 215).
evoked response potentials in the brain after the adminis- Magnetic resonance imaging (MRI) has been used in a
tration of different stimuli. Abnormal evoked response po- number of studies to examine cerebral structure in patients

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 22 May 2014. at 00:09 For personal use only. No other uses without permission. . All rights reserved.
504 Endocrine Reviews, June 2008, 29(4):494 –511 Kodl and Seaquist • Cognition and Diabetes Mellitus

with type 1 and type 2 diabetes and has pretty consistently (221). Wessels et al. (222) performed a similar experiment but
found the brains of such subjects to have leukoariosis, which compared patients with type 1 diabetes and retinopathy,
are hyperintense white matter lesions (207, 216). One study patients with type 1 diabetes and no retinopathy, and con-
found that 69% of middle-aged adults with long-standing trols. They found that patients with type 1 diabetes and
type 1 diabetes had abnormal MRI scans, compared with 12% proliferative retinopathy had decreased gray matter density
of healthy, aged-matched volunteers, with an increased in the right inferior gyrus and right occipital lobe compared
number of larger, high-signal lesions in the cerebrum, cer- with those patients with diabetes and no retinopathy, as well
ebellum, and brain stem being the primary abnormality iden- as to controls. More recently, Wessels and colleagues applied
tified (207). However, this was not confirmed by a more voxel-based morphometry to white matter volumes. They
recent published study in which relatively young patients found that those subjects with type 1 diabetes and prolifer-
(25– 40 yr old) with type 1 diabetes for more than 15 yr did ative retinopathy had significantly smaller white matter vol-
not have a significant difference in white matter hyperin- ume than subjects without diabetes, and that smaller white
tensities compared with healthy controls. In addition, white matter volume correlated with worse performances on at-
matter hyperintensities did not correlate with depressive tention, speed of information processing, and executive func-
history, retinopathy, severe hypoglycemia, glycemia control, tion. This was not seen in patients with type 1 diabetes who
and most neurocognitive tests (with the exception of delayed had no proliferative retinopathy, suggesting a possible com-
memory) (7). This is in agreement with previous studies mon mechanism in the development of retinopathy and ce-
(3, 217). The reason for the discrepancy may have been that rebral dysfunction (6). Preliminary findings by Perantie et al.
subjects in the former study had more severe microvascular (223) have found that a history of severe hypoglycemia in
complications and that differences in cardiovascular risk fac- children is associated with decreased gray matter in the left
tors between subjects with diabetes and controls were not superior temporal region, whereas increased HbAlc levels
controlled for. In patients with type 2 diabetes, these white are associated with reductions of gray matter in the right
matter hyperintensities have been noted to correlate with cuneus and precuneus regions, reductions of white matter in
reduced performance on tests of attention, executive func- the right posterior parietal region, and increased gray matter
tion, information processing speed, and memory (216, 218). in the right prefrontal cortex.
The nature of these white matter lesions is uncertain, but Functional MRI (fMRI) has also been used to assess cere-
investigators have hypothesized that they could represent bral function in patients with diabetes. fMRI is based on the
demyelination, increased water content, angionecrosis, cys- fact that increases in cerebral blood flow and metabolism
tic infarcts, or gliosis (i.e. brain tissue scarring) (92). during stimulus-induced neuronal activation are accompa-
MRI has also demonstrated that subjects with type 2 di- nied by a relative reduction in deoxyhemoglobin content of
abetes have hippocampal and amygdala atrophy relative to the activated tissue relative to the adjacent unactivated brain.
control subjects (219). The hippocampus and amygdala are Because deoxyhemoglobin is a paramagnetic molecule, it can
responsible for such functions as memory and behavior and, be visualized by MRI. Rosenthal et al. (224) applied fMRI to
interestingly, are also found to be atrophied in Alzheimer’s the study of cerebral function during standard neurocogni-
patients (219). However, a similar study in subjects with type tive testing in subjects with type 1 diabetes subjected to both
1 diabetes failed to identify reductions in hippocampal and euglycemia and hypoglycemia. They found that the effect of
amygdala volume, although these subjects did have an in- acute hypoglycemia on cerebral blood flow is task and region
crease in cerebrospinal fluid on MRI, suggesting mild global specific. For example, during hypoglycemia, the slower fin-
cerebral atrophy (103). Another study compared MRI find- ger tapping corresponded to decreased activation of the right
ings and neuropsychometric testing in patients with an early premotor cortex, supplementary motor area, and left hip-
age of onset of type 1 diabetes (younger than age 7) and a later pocampus and with increased activation in the left cerebel-
age of onset (7–17 yr old). Subjects with early onset disease lum and right frontal pole. In addition, during hypoglycemia
had larger ventricular volumes and more prevalent ventric- deterioration of four-choice reaction time correlated with
ular atrophy than those with later onset, which corresponded reduced activation in the motor and visual systems but with
to poorer intellectual and information processing ability (220). increased activation of the part of the parietal cortex involved
Atrophy in subcortical and periventricular areas has also been in planning (224). More recently, Wessels et al. (24) applied
associated with reduced performance on memory tasks in pa- this methodology to determine whether subjects with type 1
tients with type 2 diabetes (216). A history of hypoglycemia diabetes with known hyperglycemia-induced end organ
appears to be related to an increase in cerebral atrophy (70). damage, specifically retinopathy, had differing areas of ce-
Recently, voxel-based morphometry, in which differences rebral activation with cognitive stimuli and hypoglycemia
in local characteristics of tissue are measured using MRI, has compared with patients with type 1 diabetes and no micro-
been used to evaluate both the gray and white matter of vascular complications. Although there was no difference
patients with type 1 diabetes. Musen et al. (221) found that seen in cognitive ability between the two groups, there was
compared with controls, patients with type 1 diabetes had an overall increase in activation and less appropriate deac-
lower gray matter density in certain areas of the temporal tivation of certain brain regions during hypoglycemia in the
lobe, frontal lobe, and thalamus. They also found that higher diabetic group with retinopathy. The investigators hypoth-
HbA1c levels correlated with lower gray matter density in esized that regional alterations in activation were secondary
areas important for language, memory, and attention, to hyperglycemia-induced end organ damage in the central
whereas a history of severe hypoglycemia correlated with nervous system, causing altered neurovascular coupling or
less gray matter density in the left cerebellar posterior lobe functional microvascular alterations (24). Preliminary find-

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 22 May 2014. at 00:09 For personal use only. No other uses without permission. . All rights reserved.
Kodl and Seaquist • Cognition and Diabetes Mellitus Endocrine Reviews, June 2008, 29(4):494 –511 505

ings by Musen et al. (225) have found decreased activation in cursor to true dementia or represent another process. We also
the superior temporal gyrus and the parahippocampal gyrus do not know whether the incidence of dementia is increased
in response to lexical and recognition stimuli, respectively, in in patients with type 1 diabetes compared with the rest of the
a group composed of patients with long-standing type 1 population. In prior decades, this was not an issue because
diabetes compared with controls. patients with type 1 diabetes died at relatively young ages
Other imaging modalities such as single photon emission from other complications of the disease. However, now that
computed tomography (SPECT) and PET have been used to patients with type 1 diabetes are living longer and better with
assess cerebral function in patients with diabetes mellitus. the disease, this must be assessed. Finally, it is not clear
SPECT is particularly good at assessing cerebral perfusion whether the subtle cognitive deficits identified in many stud-
and has demonstrated in an uncontrolled study that patients ies truly impact the lives of patients living with diabetes (34).
with type 2 diabetes and dementia have a high incidence of Future studies, perhaps longitudinal in nature or involving
hypoperfusion in at least one area of the brain (226). How- better serum/imaging biomarkers, will be of tremendous
ever, SPECT has also demonstrated that patients with type benefit in providing better understanding of the natural his-
1 diabetes have hyperperfusion in the prefrontal and frontal tory of this complication.
brain regions compared with controls (227). Other investi- Future study will also be important in understanding the
gators found that when the SPECT data are corrected for the pathogenesis of cognitive dysfunction secondary to diabetes.
increase in cerebral atrophy seen in patients with diabetes, Although it seems that hyperglycemia and hyperglycemia-
cerebral blood flow and glucose metabolism values were induced end organ damage contribute to this problem, the
within normal range (228). PET with fluorodeoxyglucose is actual mechanisms through which hyperglycemia alters ce-
a technique that can be used to assess glucose metabolism rebral structure and function are not clear. Improved glyce-
because the compound is taken up and trapped in the cell by mic control is likely of therapeutic benefit, as has been sug-
phosphorylation. When this method was used in patients gested by many retrospective studies (5, 15–18, 26 –28, 48),
with type 1 diabetes and a history of severe hypoglycemia but a prospective study is needed to determine whether this
and hypoglycemia unawareness, no differences in glucose is true. In addition, identification of the mechanisms through
metabolism were found relative to controls, although neu- which hyperglycemia may impair cognitive function in pa-
ropsychological testing was also not significantly different tients with diabetes will stimulate new research into ways to
(69). Two pilot studies, one by our own laboratory, have prevent and treat all of the hyperglycemia-associated com-
used diffusion tensor imaging, a type of MRI that mea- plications of diabetes.
sures white matter integrity quantitatively by fractional
anisotropy (229, 230), in patients with diabetes. Prelimi-
nary findings show a reduction in white matter integrity VI. Conclusion
in patients with type 1 diabetes that was associated with In conclusion, there have been significant gains in our
severity of hyperglycemia (231) and poorer performance understanding of the effect of diabetes on cognitive dys-
on certain neurocognitive tests (67). function. Evidence from neurocognitive testing suggests that
In general, assessment modalities to detect cognitive dys- cognitive dysfunction should be listed as one of the many
function associated with diabetes have been disappointing. complications of diabetes, along with retinopathy, neurop-
Neurocognitive testing is cumbersome and lacks pathophys- athy, nephropathy, and cardiovascular disease. The patho-
iological insight. Evoked response potentials and EEG seem genesis of cognitive dysfunction is only partially understood.
to be good at detecting sensory/perception deficits but are Although many studies suggest that changes in cerebral
also cumbersome and do not provide as much information structure and function in diabetes are related to hypergly-
about more complex cognitive functions. Although promis- cemia-induced end organ damage, macrovascular disease,
ing, there have been conflicting results with regard to MRI hypoglycemia, insulin resistance, and amyloid lesions may
studies, specifically in patients with type 1 diabetes. The play a role in some patients. Greater understanding of the
utility of SPECT, PET, and diffusion tensor imaging in mon- natural history of this diabetes complication and the mech-
itoring or detecting changes in patients with cognitive dys- anisms responsible for its development will continue to ad-
function and diabetes remains to be determined. vance as biochemical and imaging modalities continue to
evolve. As new knowledge is gained, it can be applied to
develop new and improved ways to prevent and treat all of
V. Future Directions the hyperglycemia-related complications of diabetes.
Although much research has been done on the impact of
diabetes mellitus on cognitive function, many questions still Acknowledgments
remain. It is clear that patients with type 1 and type 2 diabetes
have been found to have abnormalities in neurocognitive Received October 1, 2007. Accepted March 28, 2008.
function, although the natural history and clinical signifi- Address all correspondence and requests for reprints to: Elizabeth R.
cance of these findings have not yet been clearly defined. For Seaquist, M. D., Department of Medicine, Division of Endocrinology and
example, we know that there is an increased incidence of Diabetes, University of Minnesota, Campus Delivery Code 1932, Suite
229, 717 Delaware Street SE, Minneapolis, Minnesota 55414. E-mail:
dementia in patients with type 2 diabetes (37, 38, 40 – 44), but seaqu001@umn.edu
we do not know whether the more subtle changes in memory This work was supported by NIH Grants NS35192 (to E.R.S.),
and in other measures on neurocognitive testing are a pre- DK62440 (to E.R.S.), and 2 T32 DK007203–29A1 (to C.T.K.).

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 22 May 2014. at 00:09 For personal use only. No other uses without permission. . All rights reserved.
506 Endocrine Reviews, June 2008, 29(4):494 –511 Kodl and Seaquist • Cognition and Diabetes Mellitus

Disclosure Statement: E.R.S. has served on advisory boards for Pfizer alters mood state and impairs cognitive performance in people with
and Merck. C.T.K. has no disclosures to state. type 2 diabetes. Diabetes Care 27:2335–2340
22. Brands AM, Biessels GJ, de Haan EH, Kappelle LJ, Kessels RP
2005 The effects of type 1 diabetes on cognitive performance: a
References meta-analysis. Diabetes Care 28:726 –735
23. Ferguson SC, Blane A, Perros P, McCrimmon RJ, Best JJ, Wardlaw
1. 2003 U.S. Renal Data System (USRDS) Annual Data Report: Atlas J, Deary IJ, Frier BM 2003 Cognitive ability and brain structure in
of end-stage renal disease in the United States. Bethesda, MD: type 1 diabetes: relation to microangiopathy and preceding severe
National Institutes of Health, National Institute of Diabetes and hypoglycemia. Diabetes 52:149 –156
Digestive and Kidney Diseases 24. Wessels AM, Rombouts SA, Simsek S, Kuijer JP, Kostense PJ,
2. Miles WR, Root HF 1922 Psychologic tests applied to diabetic Barkhof F, Scheltens P, Snoek FJ, Heine RJ 2006 Microvascular
patients. Arch Intern Med 30:767–777 disease in type 1 diabetes alters brain activation: a functional mag-
3. Brands AM, Kessels RP, Hoogma RP, Henselmans JM, van der netic resonance imaging study. Diabetes 55:334 –340
Beek Boter JW, Kappelle LJ, de Haan EH, Biessels GJ 2006 Cog- 25. Gregg EW, Yaffe K, Cauley JA, Rolka DB, Blackwell TL, Narayan
nitive performance, psychological well-being, and brain magnetic KM, Cummings SR 2000 Is diabetes associated with cognitive
resonance imaging in older patients with type 1 diabetes. Diabetes impairment and cognitive decline among older women? Study of
55:1800 –1806 Osteoporotic Fractures Research Group. Arch Intern Med 160:
4. Ryan CM, Geckle MO, Orchard TJ 2003 Cognitive efficiency de- 174 –180
clines over time in adults with type 1 diabetes: effects of micro- and 26. Reaven GM, Thompson LW, Nahum D, Haskins E 1990 Rela-
macrovascular complications. Diabetologia 46:940 –948 tionship between hyperglycemia and cognitive function in older
5. Ryan CM, Williams TM, Finegold DN, Orchard TJ 1993 Cognitive NIDDM patients. Diabetes Care 13:16 –21
dysfunction in adults with type 1 (insulin-dependent) diabetes 27. Munshi M, Grande L, Hayes M, Ayres D, Suhl E, Capelson R, Lin
mellitus of long duration: effects of recurrent hypoglycaemia and S, Milberg W, Weinger K 2006 Cognitive dysfunction is associated
other chronic complications. Diabetologia 36:329 –334 with poor diabetes control in older adults. Diabetes Care 29:1794 –
6. Wessels AM, Rombouts SA, Remijnse PL, Boom Y, Scheltens P, 1799
Barkhof F, Heine RJ, Snoek FJ 2007 Cognitive performance in type 28. Perlmuter LC, Hakami MK, Hodgson-Harrington C, Ginsberg J,
1 diabetes patients is associated with cerebral white matter volume. Katz J, Singer DE, Nathan DM 1984 Decreased cognitive function
Diabetologia 20:1763–1769 in aging non-insulin-dependent diabetic patients. Am J Med 77:
7. Weinger K, Jacobson AM, Musen G, Lyoo IK, Ryan CM, Jimerson 1043–1048
DC, Renshaw PF 2008 The effects of type 1 diabetes on cerebral 29. Messier C 2005 Impact of impaired glucose tolerance and type 2
white matter. Diabetologia 51:417– 425 diabetes on cognitive aging. Neurobiol Aging 26(Suppl 1):26 –30
8. Hershey T, Bhargava N, Sadler M, White NH, Craft S 1999 Con- 30. Grodstein F, Chen J, Wilson RS, Manson JE 2001 Type 2 diabetes
ventional versus intensive diabetes therapy in children with type and cognitive function in community-dwelling elderly women.
1 diabetes: effects on memory and motor speed. Diabetes Care Diabetes Care 24:1060 –1065
22:1318 –1324 31. Kanaya AM, Barrett-Connor E, Gildengorin G, Yaffe K 2004
9. Ryan CM 1988 Neurobehavioral complications of type I diabetes. Change in cognitive function by glucose tolerance status in older
Examination of possible risk factors. Diabetes Care 11:86 –93 adults: a 4-year prospective study of the Rancho Bernardo study
10. Skenazy JA, Bigler ED 1984 Neuropsychological findings in dia- cohort. Arch Intern Med 164:1327–1333
betes mellitus. J Clin Psychol 40:246 –258
32. Mooradian AD, Perryman K, Fitten J, Kavonian GD, Morley JE
11. Hershey T, Craft S, Bhargava N, White NH 1997 Memory and
1988 Cortical function in elderly non-insulin dependent diabetic
insulin dependent diabetes mellitus (IDDM): effects of childhood
patients. Behavioral and electrophysiologic studies. Arch Intern
onset and severe hypoglycemia. J Int Neuropsychol Soc 3:509 –520
Med 148:2369 –2372
12. Northam EA, Anderson PJ, Werther GA, Warne GL, Adler RG,
33. Fontbonne A, Berr C, Ducimetiere P, Alperovitch A 2001 Changes
Andrewes D 1998 Neuropsychological complications of IDDM in
in cognitive abilities over a 4-year period are unfavorably affected
children 2 years after disease onset. Diabetes Care 21:379 –384
13. Schoenle EJ, Schoenle D, Molinari L, Largo RH 2002 Impaired in elderly diabetic subjects: results of the Epidemiology of Vascular
intellectual development in children with type I diabetes: associ- Aging Study. Diabetes Care 24:366 –370
ation with HbA(1c), age at diagnosis and sex. Diabetologia 45: 34. Sinclair AJ, Girling AJ, Bayer AJ 2000 Cognitive dysfunction in
108 –114 older subjects with diabetes mellitus: impact on diabetes self-
14. Northam EA, Anderson PJ, Jacobs R, Hughes M, Warne GL, management and use of care services. All Wales Research into
Werther GA 2001 Neuropsychological profiles of children with Elderly (AWARE) Study. Diabetes Res Clin Pract 50:203–212
type 1 diabetes 6 years after disease onset. Diabetes Care 24:1541– 35. Gregg EW, Brown AM 2003 Cognitive and physical disabilities and
1546 aging-related complications of diabetes. Clin Diabetes 21:113–118
15. Jacobson AM, Musen G, Ryan CM, Silvers N, Cleary P, Waberski 36. Bruce DG, Casey GP, Grange V, Clarnette RC, Almeida OP,
B, Burwood A, Weinger K, Bayless M, Dahms W, Harth J 2007 Foster JK, Ives FJ, Davis TM 2003 Cognitive impairment, physical
Long-term effect of diabetes and its treatment on cognitive func- disability and depressive symptoms in older diabetic patients: the
tion. N Engl J Med 356:1842–1852 Fremantle Cognition in Diabetes Study. Diabetes Res Clin Pract
16. Holmes CS 1986 Neuropsychological profiles in men with insulin- 61:59 – 67
dependent diabetes. J Consult Clin Psychol 54:386 –389 37. Cukierman T, Gerstein HC, Williamson JD 2005 Cognitive decline
17. Kaufman FR, Epport K, Engilman R, Halvorson M 1999 Neuro- and dementia in diabetes–systematic overview of prospective ob-
cognitive functioning in children diagnosed with diabetes before servational studies. Diabetologia 48:2460 –2469
age 10 years. J Diabetes Complications 13:31–38 38. Curb JD, Rodriguez BL, Abbott RD, Petrovitch H, Ross GW,
18. Perantie DC, Lim A, Wu J, Weaver P, Warren SL, Hayden T, Christ Masaki KH, Foley D, Blanchette PL, Harris T, Chen R, White LR
S, Sadler M, White NH, Hershey T 2007 Severe hypoglycemia vs. 1999 Longitudinal association of vascular and Alzheimer’s demen-
hyperglycemia: unique effects on cognition in youth with type 1 tias, diabetes, and glucose tolerance. Neurology 52:971–975
diabetes mellitus (TIDM). Diabetes 56(Suppl 1):(Abstract 1887) 39. Kuusisto J, Koivisto K, Mykkanen L, Helkala EL, Vanhanen M,
19. Cox DJ, Kovatchev BP, Gonder-Frederick LA, Summers KH, Hanninen T, Kervinen K, Kesaniemi YA, Riekkinen PJ, Laakso
McCall A, Grimm KJ, Clarke WL 2005 Relationships between M 1997 Association between features of the insulin resistance syn-
hyperglycemia and cognitive performance among adults with type drome and Alzheimer’s disease independently of apolipoprotein
1 and type 2 diabetes. Diabetes Care 28:71–77 E4 phenotype: cross-sectional population based study. BMJ 315:
20. Rovet J, Alvarez M 1997 Attentional functioning in children and 1045–1049
adolescents with IDDM. Diabetes Care 20:803– 810 40. Leibson CL, Rocca WA, Hanson VA, Cha R, Kokmen E, O’Brien
21. Sommerfield AJ, Deary IJ, Frier BM 2004 Acute hyperglycemia PC, Palumbo PJ 1997 Risk of dementia among persons with dia-

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 22 May 2014. at 00:09 For personal use only. No other uses without permission. . All rights reserved.
Kodl and Seaquist • Cognition and Diabetes Mellitus Endocrine Reviews, June 2008, 29(4):494 –511 507

betes mellitus: a population-based cohort study. Am J Epidemiol 60. Sommerfield AJ, Deary IJ, McAulay V, Frier BM 2003 Short-term,
145:301–308 delayed, and working memory are impaired during hypoglycemia
41. Luchsinger JA, Tang MX, Stern Y, Shea S, Mayeux R 2001 Dia- in individuals with type 1 diabetes. Diabetes Care 26:390 –396
betes mellitus and risk of Alzheimer’s disease and dementia with 61. Widom B, Simonson DC 1990 Glycemic control and neuropsy-
stroke in a multiethnic cohort. Am J Epidemiol 154:635– 641 chologic function during hypoglycemia in patients with insulin-
42. Ott A, Stolk RP, Hofman A, van Harskamp F, Grobbee DE, dependent diabetes mellitus. Ann Intern Med 112:904 –912
Breteler MM 1996 Association of diabetes mellitus and dementia: 62. Warren RE, Zammitt NN, Deary IJ, Frier BM 2007 The effects of
the Rotterdam Study. Diabetologia 39:1392–1397 acute hypoglycaemia on memory acquisition and recall and pro-
43. Peila R, Rodriguez BL, Launer LJ 2002 Type 2 diabetes, APOE spective memory in type 1 diabetes. Diabetologia 50:178 –185
gene, and the risk for dementia and related pathologies: the 63. Maran A, Lomas J, Macdonald IA, Amiel SA 1995 Lack of pres-
Honolulu-Asia Aging Study. Diabetes 51:1256 –1262 ervation of higher brain function during hypoglycaemia in patients
44. Sullivan MG, Glycemic control shown to prevent dementia. 10th with intensively-treated IDDM. Diabetologia 38:1412–1418
International Conference on Alzheimer’s Disease and Related Dis- 64. Deary IJ, Crawford JR, Hepburn DA, Langan SJ, Blackmore LM,
orders, Madrid, Spain, 2006 Frier BM 1993 Severe hypoglycemia and intelligence in adult pa-
45. Yoshitake T, Kiyohara Y, Kato I, Ohmura T, Iwamoto H, Nakayama K, tients with insulin-treated diabetes. Diabetes 42:341–344
Ohmori S, Nomiyama K, Kawano H, Ueda K, Sueshi K, Tsuneyoshi 65. Howorka K, Pumprla J, Saletu B, Anderer P, Krieger M, Schabmann
M, Fujishima M 1995 Incidence and risk factors of vascular dementia and A 2000 Decrease of vigilance assessed by EEG-mapping in type I
Alzheimer’s disease in a defined elderly Japanese population: the Hi- diabetic patients with history of recurrent severe hypoglycaemia.
sayama Study. Neurology 45:1161–1168 Psychoneuroendocrinology 25:85–105
46. Watson GS, Craft S 2003 The role of insulin resistance in the 66. Wredling R, Levander S, Adamson U, Lins PE 1990 Permanent
pathogenesis of Alzheimer’s disease: implications for treatment. neuropsychological impairment after recurrent episodes of severe
CNS Drugs 17:27– 45 hypoglycaemia in man. Diabetologia 33:152–157
47. Yaffe K, Blackwell T, Whitmer RA, Krueger K, Barrett Connor E 67. Kodl C, Franc D, Anderson FS, Lim KO, Seaquist ER, Diffusion
2006 Glycosylated hemoglobin level and development of mild cog- tensor imaging (DTI) identifies defects in white matter microstruc-
nitive impairment or dementia in older women. J Nutr Health ture in subjects with diabetes. American Diabetes Association 67th
Aging 10:293–295 Scientific Session, Chicago, IL, 2007
48. Ryan CM, Geckle MO 2000 Circumscribed cognitive dysfunction 68. DCCT Research Group 1996 Effects of intensive diabetes therapy
in middle-aged adults with type 2 diabetes. Diabetes Care 23:1486 – on neuropsychological function in adults in the Diabetes Control
1493 and Complications Trial. Ann Intern Med 124:379 –388
49. Vanhanen M, Koivisto K, Kuusisto J, Mykkanen L, Helkala EL, 69. Chabriat H, Sachon C, Levasseur M, Grimaldi A, Pappata S,
Hanninen T, Riekkinen Sr P, Soininen H, Laakso M 1998 Cog- Rougemont D, Masure MC, De Recondo A, Samson Y 1994 Brain
nitive function in an elderly population with persistent impaired metabolism after recurrent insulin induced hypoglycaemic epi-
glucose tolerance. Diabetes Care 21:398 – 402 sodes: a PET study. J Neurol Neurosurg Psychiatry 57:1360 –1365
50. de Vegt F, Dekker JM, Ruhe HG, Stehouwer CD, Nijpels G, 70. Perros P, Deary IJ, Sellar RJ, Best JJ, Frier BM 1997 Brain abnor-
Bouter LM, Heine RJ 1999 Hyperglycaemia is associated with malities demonstrated by magnetic resonance imaging in adult
all-cause and cardiovascular mortality in the Hoorn population: the IDDM patients with and without a history of recurrent severe
Hoorn Study. Diabetologia 42:926 –931 hypoglycemia. Diabetes Care 20:1013–1018
51. Dilley J, Ganesan A, Deepa R, Deepa M, Sharada G, Williams 71. Reichard P, Britz A, Rosenqvist U 1991 Intensified conventional
OD, Mohan V 2007 Association of A1C with cardiovascular disease insulin treatment and neuropsychological impairment. BMJ 303:
and metabolic syndrome in Asian Indians with normal glucose 1439 –1442
tolerance. Diabetes Care 30:1527–1532 72. Ack M, Miller I, Weil Jr WB 1961 Intelligence of children with
52. Khaw KT, Wareham N, Bingham S, Luben R, Welch A, Day N diabetes mellitus. Pediatrics 28:764 –770
2004 Association of hemoglobin A1c with cardiovascular disease 73. Ryan C, Vega A, Drash A 1985 Cognitive deficits in adolescents
and mortality in adults: the European prospective investigation who developed diabetes early in life. Pediatrics 75:921–927
into cancer in Norfolk. Ann Intern Med 141:413– 420 74. Ryan CM 1997 Effects of diabetes mellitus on neuropsychological
53. Kumari M, Marmot M 2005 Diabetes and cognitive function in a functioning: a lifespan perspective. Semin Clin Neuropsychiatry
middle-aged cohort: findings from the Whitehall II study. Neu- 2:4 –14
rology 65:1597–1603 75. Biessels GJ, van der Heide LP, Kamal A, Bleys RL, Gispen WH
54. Scott RD, Kritz-Silverstein D, Barrett-Connor E, Wiederholt WC 2002 Ageing and diabetes: implications for brain function. Eur
1998 The association of non-insulin-dependent diabetes mellitus J Pharmacol 441:1–14
and cognitive function in an older cohort. J Am Geriatr Soc 46: 76. Brownlee M 2005 The pathobiology of diabetic complications: a
1217–1222 unifying mechanism. Diabetes 54:1615–1625
55. Lindeman RD, Romero LJ, LaRue A, Yau CL, Schade DS, Koehler 77. Klein JP, Waxman SG 2003 The brain in diabetes: molecular
KM, Baumgartner RN, Garry PJ 2001 A biethnic community sur- changes in neurons and their implications for end-organ damage.
vey of cognition in participants with type 2 diabetes, impaired Lancet Neurol 2:548 –554
glucose tolerance, and normal glucose tolerance: the New Mexico 78. Toth C, Schmidt AM, Tuor UI, Francis G, Foniok T, Brussee V,
Elder Health Survey. Diabetes Care 24:1567–1572 Kaur J, Yan SF, Martinez JA, Barber PA, Buchan A, Zochodne DW
56. 1993 The effect of intensive treatment of diabetes on the develop- 2006 Diabetes, leukoencephalopathy and rage. Neurobiol Dis 23:
ment and progression of long-term complications in insulin- 445– 461
dependent diabetes mellitus. The Diabetes Control and Compli- 79. Sredy J, Sawicki DR, Notvest RR 1991 Polyol pathway activity in
cations Trial Research Group. N Engl J Med 329:977–986 nervous tissues of diabetic and galactose-fed rats: effect of dietary
57. 1998 Intensive blood-glucose control with sulphonylureas or in- galactose withdrawal or tolrestat intervention therapy. J Diabet
sulin compared with conventional treatment and risk of compli- Complications 5:42– 47
cations in patients with type 2 diabetes (UKPDS 33). UK Prospec- 80. Malone JI, Hanna SK, Liang X, Saporta S, Park CR, Diamond D
tive Diabetes Study (UKPDS) Group. Lancet 352:837– 853 2007 Diabetes increases polyol pathway activity in the brain which
58. Nathan DM, Cleary PA, Backlund JY, Genuth SM, Lachin JM, is blocked by sorbinil. Diabetes 56(Suppl 1) (Abstract 793)
Orchard TJ, Raskin P, Zinman B 2005 Intensive diabetes treatment 81. Yan SD, Stern D, Schmidt AM 1997 What’s the RAGE? The re-
and cardiovascular disease in patients with type 1 diabetes. N Engl ceptor for advanced glycation end products (RAGE) and the dark
J Med 353:2643–2653 side of glucose. Eur J Clin Invest 27:179 –181
59. Austin EJ, Deary IJ 1999 Effects of repeated hypoglycemia on 82. Girones X, Guimera A, Cruz-Sanchez CZ, Ortega A, Sasaki N,
cognitive function: a psychometrically validated reanalysis of the Makita Z, Lafuente JV, Kalaria R, Cruz-Sanchez FF 2004 N ␧-
Diabetes Control and Complications Trial data. Diabetes Care 22: carboxymethyllysine in brain aging, diabetes mellitus, and
1273–1277 Alzheimer’s disease. Free Radic Biol Med 36:1241–1247

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 22 May 2014. at 00:09 For personal use only. No other uses without permission. . All rights reserved.
508 Endocrine Reviews, June 2008, 29(4):494 –511 Kodl and Seaquist • Cognition and Diabetes Mellitus

83. Heitner J, Dickson D 1997 Diabetics do not have increased Al- treated diabetes mellitus: clinical features and neuropathological
zheimer-type pathology compared with age-matched control sub- changes. Diabet Med 7:349 –354
jects. A retrospective postmortem immunocytochemical and hist- 105. Auer RN 2004 Hypoglycemic brain damage. Metab Brain Dis 19:
ofluorescent study. Neurology 49:1306 –1311 169 –175
84. Niiya Y, Abumiya T, Shichinohe H, Kuroda S, Kikuchi S, Ieko M, 106. Auer RN 2004 Hypoglycemic brain damage. Forensic Sci Int 146:
Yamagishi S, Takeuchi M, Sato T, Iwasaki Y 2006 Susceptibility 105–110
of brain microvascular endothelial cells to advanced glycation end 107. Elusiyan JB, Adeodu OO, Adejuyigbe EA 2006 Evaluating the
products-induced tissue factor upregulation is associated with in- validity of a bedside method of detecting hypoglycemia in children.
tracellular reactive oxygen species. Brain Res 1108:179 –187 Pediatr Emerg Care 22:488 – 490
85. Zimmerman GA, Meistrell 3rd M, Bloom O, Cockroft KM, 108. Comi G 1997 Evoked potentials in diabetes mellitus. Clin Neurosci
Bianchi M, Risucci D, Broome J, Farmer P, Cerami A, Vlassara H, 4:374 –379
Tracey KJ 1995 Neurotoxicity of advanced glycation endproducts 109. Siesjo BK, Bengtsson F 1989 Calcium fluxes, calcium antagonists,
during focal stroke and neuroprotective effects of aminoguanidine. and calcium-related pathology in brain ischemia, hypoglycemia,
Proc Natl Acad Sci USA 92:3744 –3748 and spreading depression: a unifying hypothesis. J Cereb Blood
86. Ramakrishnan R, Sheeladevi R, Suthanthirarajan N 2004 PKC-␣ Flow Metab 9:127–140
mediated alterations of indoleamine contents in diabetic rat brain. 110. Wieloch T 1985 Hypoglycemia-induced neuronal damage pre-
Brain Res Bull 64:189 –194 vented by an N-methyl-D-aspartate antagonist. Science 230:681–
87. Sieber FE, Hurn P, Alkayed NJ, Traystman RJ 2001 Gender-based 683
differences in Na⫹ -K⫹ adenosine triphosphatase activity occur in 111. Jauch-Chara K, Hallschmid M, Gais S, Schmid SM, Oltmanns
the microcirculation of the diabetic rat brain. Anesthesiology 94: KM, Colmorgen C, Born J, Schultes B 2007 Hypoglycemia during
372–375 sleep impairs consolidation of declarative memory in type 1 dia-
88. Castellani RJ, Siedlak SL, Fortino AE, Perry G, Ghetti B, Smith betic and healthy humans. Diabetes Care 30:2040 –2045
MA 2005 Chitin-like polysaccharides in Alzheimer’s disease brains. 112. Bendtson I, Gade J, Theilgaard A, Binder C 1992 Cognitive func-
Curr Alzheimer Res 2:419 – 423 tion in type 1 (insulin-dependent) diabetic patients after nocturnal
89. Aragno M, Mastrocola R, Medana C, Restivo F, Catalano MG, hypoglycaemia. Diabetologia 35:898 –903
Pons N, Danni O, Boccuzzi G 2005 Up-regulation of advanced 113. Matyka KA, Wigg L, Pramming S, Stores G, Dunger DB 1999
glycated products receptors in the brain of diabetic rats is pre- Cognitive function and mood after profound nocturnal hypogly-
vented by antioxidant treatment. Endocrinology 146:5561–5567 caemia in prepubertal children with conventional insulin treatment
90. Kamal A, Biessels GJ, Urban IJ, Gispen WH 1999 Hippocampal for diabetes. Arch Dis Child 81:138 –142
synaptic plasticity in streptozotocin-diabetic rats: impairment of 114. Marks JL, Porte Jr D, Stahl WL, Baskin DG 1990 Localization of
long-term potentiation and facilitation of long-term depression. insulin receptor mRNA in rat brain by in situ hybridization. En-
Neuroscience 90:737–745 docrinology 127:3234 –3236
91. Welsh B, Wecker L 1991 Effects of streptozotocin-induced diabetes 115. Unger J, McNeill TH, Moxley 3rd RT, White M, Moss A,
on acetylcholine metabolism in rat brain. Neurochem Res 16: Livingston JN 1989 Distribution of insulin receptor-like immuno-
453– 460 reactivity in the rat forebrain. Neuroscience 31:143–157
92. Biessels GJ, Kappelle AC, Bravenboer B, Erkelens DW, Gispen 116. Brant AM, Jess TJ, Milligan G, Brown CM, Gould GW 1993
WH 1994 Cerebral function in diabetes mellitus. Diabetologia 37: Immunological analysis of glucose transporters expressed in dif-
643– 650 ferent regions of the rat brain and central nervous system. Biochem
93. McCall AL 1992 The impact of diabetes on the CNS. Diabetes Biophys Res Commun 192:1297–1302
41:557–570 117. Ibberson M, Uldry M, Thorens B 2000 GLUTX1, a novel mam-
94. Reske-Nielsen E, Lundbaek K 1965 Pathological changes in the malian glucose transporter expressed in the central nervous system
central and peripheral nervous system of young long-term diabet- and insulin-sensitive tissues. J Biol Chem 275:4607– 4612
ics. I. Diabetic encephalopathy. Diabetologia 1:233–241 118. Carayannopoulos MO, Chi MM, Cui Y, Pingsterhaus JM,
95. Reske-Nielsen E, Lundbaek K 1968 Pathological changes in the McKnight RA, Mueckler M, Devaskar SU, Moley KH 2000
central and peripheral nervous system of young long-term diabet- GLUT8 is a glucose transporter responsible for insulin-stimulated
ics. II. The spinal cord and peripheral nerves. Diabetologia 4:34 – 43 glucose uptake in the blastocyst. Proc Natl Acad Sci USA 97:7313–
96. Johnson PC, Brendel K, Meezan E 1982 Thickened cerebral cortical 7318
capillary basement membranes in diabetics. Arch Pathol Lab Med 119. Reagan LP, Gorovits N, Hoskin EK, Alves SE, Katz EB, Grillo CA,
106:214 –217 Piroli GG, McEwen BS, Charron MJ 2001 Localization and reg-
97. Rodriguez G, Nobili F, Celestino MA, Francione S, Gulli G, ulation of GLUTx1 glucose transporter in the hippocampus of
Hassan K, Marenco S, Rosadini G, Cordera R 1993 Regional ce- streptozotocin diabetic rats. Proc Natl Acad Sci USA 98:2820 –2825
rebral blood flow and cerebrovascular reactivity in IDDM. Diabetes 120. Seaquist ER, Damberg GS, Tkac I, Gruetter R 2001 The effect of
Care 16:462– 468 insulin on in vivo cerebral glucose concentrations and rates of
98. Duckrow RB, Beard DC, Brennan RW 1987 Regional cerebral glucose transport/metabolism in humans. Diabetes 50:2203–2209
blood flow decreases during chronic and acute hyperglycemia. 121. Hasselbalch SG, Knudsen GM, Videbaek C, Pinborg LH,
Stroke 18:52–58 Schmidt JF, Holm S, Paulson OB 1999 No effect of insulin on
99. Kushner M, Nencini P, Reivich M, Rango M, Jamieson D, Fazekas glucose blood-brain barrier transport and cerebral metabolism in
F, Zimmerman R, Chawluk J, Alavi A, Alves W 1990 Relation of humans. Diabetes 48:1915–1921
hyperglycemia early in ischemic brain infarction to cerebral anat- 122. Namba H, Lucignani G, Nehlig A, Patlak C, Pettigrew K,
omy, metabolism, and clinical outcome. Ann Neurol 28:129 –135 Kennedy C, Sokoloff L 1987 Effects of insulin on hexose transport
100. Li PA, Shuaib A, Miyashita H, He QP, Siesjo BK, Warner DS 2000 across blood-brain barrier in normoglycemia. Am J Physiol 252:
Hyperglycemia enhances extracellular glutamate accumulation in E299 –E303
rats subjected to forebrain ischemia. Stroke 31:183–192 123. Bingham EM, Hopkins D, Smith D, Pernet A, Hallett W, Reed L,
101. Coyle JT, Puttfarcken P 1993 Oxidative stress, glutamate, and Marsden PK, Amiel SA 2002 The role of insulin in human brain
neurodegenerative disorders. Science 262:689 – 695 glucose metabolism: an 18-fluoro-deoxyglucose positron emission
102. Sima AA, Li ZG 2005 The effect of C-peptide on cognitive dys- tomography study. Diabetes 51:3384 –3390
function and hippocampal apoptosis in type 1 diabetic rats. Dia- 124. Anthony K, Reed LJ, Dunn JT, Bingham E, Hopkins D, Marsden
betes 54:1497–1505 PK, Amiel SA 2006 Attenuation of insulin-evoked responses in
103. Lognig BM, Kromeke O, Optenhostert-Porstt C, Wolf OT 2005 brain networks controlling appetite and reward in insulin resis-
Hippocampal volume and cognitive performance in long-standing tance: the cerebral basis for impaired control of food intake in
type 1 diabetic patients without macrovascular complications. Dia- metabolic syndrome? Diabetes 55:2986 –2992
bet Med 23:32–39 125. 1994 Diagnostic and statistical manual of mental disorders (DSM-IV).
104. Patrick AW, Campbell IW 1990 Fatal hypoglycaemia in insulin- Washington, DC: American Psychiatric Association

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 22 May 2014. at 00:09 For personal use only. No other uses without permission. . All rights reserved.
Kodl and Seaquist • Cognition and Diabetes Mellitus Endocrine Reviews, June 2008, 29(4):494 –511 509

126. Ferini-Strambi L, Smirne S, Garancini P, Pinto P, Franceschi M 146. Kalaria RN, Harik SI 1989 Abnormalities of the glucose transporter
1990 Clinical and epidemiological aspects of Alzheimer’s disease at the blood-brain barrier and in brain in Alzheimer’s disease. Prog
with presenile onset: a case control study. Neuroepidemiology Clin Biol Res 317:415– 421
9:39 – 49 147. Simpson IA, Chundu KR, Davies-Hill T, Honer WG, Davies P
127. Landin K, Blennow K, Wallin A, Gottfries CG 1993 Low blood 1994 Decreased concentrations of GLUT1 and GLUT3 glucose
pressure and blood glucose levels in Alzheimer’s disease. Evidence transporters in the brains of patients with Alzheimer’s disease. Ann
for a hypometabolic disorder? J Intern Med 233:357–363 Neurol 35:546 –551
128. Nielson KA, Nolan JH, Berchtold NC, Sandman CA, Mulnard 148. Benedict L, Nelson CA, Schunk E, Sullwold K, Seaquist ER 2006
RA, Cotman CW 1996 Apolipoprotein-E genotyping of diabetic Effect of insulin on the brain activity obtained during visual and
dementia patients: is diabetes rare in Alzheimer’s disease? J Am memory tasks in healthy human subjects. Neuroendocrinology
Geriatr Soc 44:897–904 83:20 –26
129. Janson J, Laedtke T, Parisi JE, O’Brien P, Petersen RC, Butler PC 149. Craft S, Asthana S, Schellenberg G, Cherrier M, Baker LD,
2004 Increased risk of type 2 diabetes in Alzheimer disease. Dia- Newcomer J, Plymate S, Latendresse S, Petrova A, Raskind M,
betes 53:474 – 481 Peskind E, Lofgreen C, Grimwood K 1999 Insulin metabolism in
130. Bucht G, Adolfsson R, Lithner F, Winblad B 1983 Changes in Alzheimer’s disease differs according to apolipoprotein E genotype
blood glucose and insulin secretion in patients with senile dementia and gender. Neuroendocrinology 70:146 –152
of Alzheimer type. Acta Med Scand 213:387–392 150. Kern W, Peters A, Fruehwald-Schultes B, Deininger E, Born J,
131. Fujisawa Y, Sasaki K, Akiyama K 1991 Increased insulin levels Fehm HL 2001 Improving influence of insulin on cognitive func-
after OGTT load in peripheral blood and cerebrospinal fluid of tions in humans. Neuroendocrinology 74:270 –280
patients with dementia of Alzheimer type. Biol Psychiatry 30:1219 – 151. Benedict C, Hallschmid M, Hatke A, Schultes B, Fehm HL, Born
1228 J, Kern W 2004 Intranasal insulin improves memory in humans.
132. Hoyer S 1998 Is sporadic Alzheimer disease the brain type of Psychoneuroendocrinology 29:1326 –1334
non-insulin dependent diabetes mellitus? A challenging hypoth- 152. Craft S, Asthana S, Cook DG, Baker LD, Cherrier M, Purganan
esis. J Neural Transm 105:415– 422 K, Wait C, Petrova A, Latendresse S, Watson GS, Newcomer JW,
133. Frolich L, Blum-Degen D, Bernstein HG, Engelsberger S, Schellenberg GD, Krohn AJ 2003 Insulin dose-response effects on
Humrich J, Laufer S, Muschner D, Thalheimer A, Turk A, Hoyer memory and plasma amyloid precursor protein in Alzheimer’s
S, Zochling R, Boissl KW, Jellinger K, Riederer P 1998 Brain disease: interactions with apolipoprotein E genotype. Psychoneu-
insulin and insulin receptors in aging and sporadic Alzheimer’s roendocrinology 28:809 – 822
disease. J Neural Transm 105:423– 438 153. Lockette W, Kirkland K, Farrow S 1996 ␣2-Adrenergic agonists
134. Craft S, Peskind E, Schwartz MW, Schellenberg GD, Raskind M, increase cellular lactate efflux. Hypertension 27:1104 –1107
Porte Jr D 1998 Cerebrospinal fluid and plasma insulin levels in 154. Holloway R, Zhou Z, Harvey HB, Levasseur JE, Rice AC, Sun D,
Alzheimer’s disease: relationship to severity of dementia and apo- Hamm RJ, Bullock MR 2007 Effect of lactate therapy upon cog-
lipoprotein E genotype. Neurology 50:164 –168 nitive deficits after traumatic brain injury in the rat. Acta Neurochir
135. Wenk GL 2006 Neuropathologic changes in Alzheimer’s disease: (Wien) 149:919 –927; discussion, 927
potential targets for treatment. J Clin Psychiatry 67(Suppl 3):3–7; 155. Maran A, Crepaldi C, Trupiani S, Lucca T, Jori E, Macdonald IA,
quiz, 23 Tiengo A, Avogaro A, Del Prato S 2000 Brain function rescue effect
136. Stone WS, Cottrill KL, Walker DL, Gold PE 1988 Blood glucose of lactate following hypoglycaemia is not an adaptation process in
and brain function: interactions with CNS cholinergic systems. both normal and type I diabetic subjects. Diabetologia 43:733–741
Behav Neural Biol 50:325–334 156. Kalman J, Palotas A, Bodi N, Kincses TZ, Benedek G, Janka Z,
137. Stone WS, Croul CE, Gold PE 1988 Attenuation of scopolamine- Antal A 2005 Lactate infusion fails to improve semantic categori-
induced amnesia in mice. Psychopharmacology (Berl) 96:417– 420 zation in Alzheimer’s disease. Brain Res Bull 65:533–539
138. Craft S, Newcomer J, Kanne S, Dagogo-Jack S, Cryer P, Sheline 157. Hak AE, Pols HA, Stehouwer CD, Meijer J, Kiliaan AJ, Hofman
Y, Luby J, Dagogo-Jack A, Alderson A 1996 Memory improvement A, Breteler MM, Witteman JC 2001 Markers of inflammation and
following induced hyperinsulinemia in Alzheimer’s disease. Neu- cellular adhesion molecules in relation to insulin resistance in non-
robiol Aging 17:123–130 diabetic elderly: the Rotterdam study. J Clin Endocrinol Metab
139. Craft S, Dagogo-Jack SE, Wiethop BV, Murphy C, Nevins RT, 86:4398 – 4405
Fleischman S, Rice V, Newcomer JW, Cryer PE 1993 Effects of 158. Hull M, Strauss S, Berger M, Volk B, Bauer J 1996 The partici-
hyperglycemia on memory and hormone levels in dementia of the pation of interleukin-6, a stress-inducible cytokine, in the patho-
Alzheimer type: a longitudinal study. Behav Neurosci 107:926 –940 genesis of Alzheimer’s disease. Behav Brain Res 78:37– 41
140. Reger MA, Watson GS, Frey 2nd WH, Baker LD, Cholerton B, 159. Rosler N, Wichart I, Jellinger KA 2001 Intra vitam lumbar and post
Keeling ML, Belongia DA, Fishel MA, Plymate SR, Schellenberg mortem ventricular cerebrospinal fluid immunoreactive interleu-
GD, Cherrier MM, Craft S 2006 Effects of intranasal insulin on kin-6 in Alzheimer’s disease patients. Acta Neurol Scand 103:126 –
cognition in memory-impaired older adults: modulation by APOE 130
genotype. Neurobiol Aging 27:451– 458 160. Tojo C, Takao T, Nishioka T, Numata Y, Suemaru S, Hashimoto
141. Skeberdis VA, Lan J, Zheng X, Zukin RS, Bennett MV 2001 K 1996 Hypothalamic-pituitary-adrenal axis in WBN/Kob rats
Insulin promotes rapid delivery of N-methyl-D- aspartate recep- with non-insulin dependent diabetes mellitus. Endocr J 43:233–239
tors to the cell surface by exocytosis. Proc Natl Acad Sci USA 161. Lee ZS, Chan JC, Yeung VT, Chow CC, Lau MS, Ko GT, Li JK,
98:3561–3566 Cockram CS, Critchley JA 1999 Plasma insulin, growth hormone,
142. Palovcik RA, Phillips MI, Kappy MS, Raizada MK 1984 Insulin cortisol, and central obesity among young Chinese type 2 diabetic
inhibits pyramidal neurons in hippocampal slices. Brain Res 309: patients. Diabetes Care 22:1450 –1457
187–191 162. Newcomer JW, Craft S, Hershey T, Askins K, Bardgett ME 1994
143. Mirzaei S, Gelpi E, Booij J, Rodrigues M, Neumann I, Zaknun J, Glucocorticoid-induced impairment in declarative memory per-
Koehn H, Knoll P 2004 New approaches in nuclear medicine for formance in adult humans. J Neurosci 14:2047–2053
early diagnosis of Alzheimer’s disease. Curr Alzheimer Res 1:219 – 163. de Quervain DJ, Roozendaal B, Nitsch RM, McGaugh JL, Hock
229 C 2000 Acute cortisone administration impairs retrieval of long-
144. Mosconi L, De Santi S, Li Y, Li J, Zhan J, Tsui WH, Boppana M, term declarative memory in humans. Nat Neurosci 3:313–314
Pupi A, de Leon MJ 2006 Visual rating of medial temporal lobe 164. Newcomer JW, Selke G, Melson AK, Hershey T, Craft S, Richards
metabolism in mild cognitive impairment and Alzheimer’s disease K, Alderson AL 1999 Decreased memory performance in healthy
using FDG-PET. Eur J Nucl Med Mol Imaging 33:210 –221 humans induced by stress-level cortisol treatment. Arch Gen Psy-
145. von Borczyskowski D, Wilke F, Martin B, Brenner W, Clausen M, chiatry 56:527–533
Mester J, Buchert R 2006 Evaluation of a new expert system for 165. Lupien S, Lecours AR, Lussier I, Schwartz G, Nair NP, Meaney
fully automated detection of the Alzheimer’s dementia pattern in MJ 1994 Basal cortisol levels and cognitive deficits in human aging.
FDG PET. Nucl Med Commun 27:739 –743 J Neurosci 14:2893–2903

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 22 May 2014. at 00:09 For personal use only. No other uses without permission. . All rights reserved.
510 Endocrine Reviews, June 2008, 29(4):494 –511 Kodl and Seaquist • Cognition and Diabetes Mellitus

166. Forget H, Lacroix A, Somma M, Cohen H 2000 Cognitive decline Purification and characterization of a peptide from amyloid-rich
in patients with Cushing’s syndrome. J Int Neuropsychol Soc pancreases of type 2 diabetic patients. Proc Natl Acad Sci USA
6:20 –29 84:8628 – 8632
167. Brunetti A, Fulham MJ, Aloj L, De Souza B, Nieman L, Oldfield 189. Lorenzo A, Razzaboni B, Weir GC, Yankner BA 1994 Pancreatic
EH, Di Chiro G 1998 Decreased brain glucose utilization in patients islet cell toxicity of amylin associated with type-2 diabetes mellitus.
with Cushing’s disease. J Nucl Med 39:786 –790 Nature 368:756 –760
168. Brown ES, Rush AJ, McEwen BS 1999 Hippocampal remodeling 190. Schubert D, Behl C, Lesley R, Brack A, Dargusch R, Sagara Y,
and damage by corticosteroids: implications for mood disorders. Kimura H 1995 Amyloid peptides are toxic via a common oxidative
Neuropsychopharmacology 21:474 – 484 mechanism. Proc Natl Acad Sci USA 92:1989 –1993
169. Gould E, Tanapat P 1999 Stress and hippocampal neurogenesis. 191. Craig EA, Weissman JS, Horwich AL 1994 Heat shock proteins and
Biol Psychiatry 46:1472–1479 molecular chaperones: mediators of protein conformation and
170. Joels M, de Kloet ER 1989 Effects of glucocorticoids and norepi- turnover in the cell. Cell 78:365–372
nephrine on the excitability in the hippocampus. Science 245:1502– 192. Li ZG, Zhang W, Sima AA 2007 Alzheimer-like changes in rat
1505 models of spontaneous diabetes. Diabetes 56:1817–1824
171. Pavlides C, McEwen BS 1999 Effects of mineralocorticoid and 193. Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE,
glucocorticoid receptors on long-term potentiation in the CA3 hip- Gaskell PC, Small GW, Roses AD, Haines JL, Pericak-Vance MA
pocampal field. Brain Res 851:204 –214 1993 Gene dose of apolipoprotein E type 4 allele and the risk of
172. Sapolsky RM, Uno H, Rebert CS, Finch CE 1990 Hippocampal Alzheimer’s disease in late onset families. Science 261:921–923
damage associated with prolonged glucocorticoid exposure in pri- 194. Watson GS, Cholerton BA, Reger MA, Baker LD, Plymate SR,
mates. J Neurosci 10:2897–2902 Asthana S, Fishel MA, Kulstad JJ, Green PS, Cook DG, Kahn SE,
173. Braak H, Braak E 1997 Frequency of stages of Alzheimer-related Keeling ML, Craft S 2005 Preserved cognition in patients with
lesions in different age categories. Neurobiol Aging 18:351–357 early Alzheimer disease and amnestic mild cognitive impairment
174. Masters CL, Multhaup G, Simms G, Pottgiesser J, Martins RN, during treatment with rosiglitazone: a preliminary study. Am J
Beyreuther K 1985 Neuronal origin of a cerebral amyloid: neuro- Geriatr Psychiatry 13:950 –958
fibrillary tangles of Alzheimer’s disease contain the same protein 195. Risner ME, Saunders AM, Altman JF, Ormandy GC, Craft S,
as the amyloid of plaque cores and blood vessels. EMBO J 4:2757– Foley IM, Zvartau-Hind ME, Hosford DA, Roses AD 2006 Efficacy
2763 of rosiglitazone in a genetically defined population with mild-to-
175. Masters CL, Simms G, Weinman NA, Multhaup G, McDonald moderate Alzheimer’s disease. Pharmacogenomics J 6:246 –254
BL, Beyreuther K 1985 Amyloid plaque core protein in Alzheimer 196. Pedersen WA, McMillan PJ, Kulstad JJ, Leverenz JB, Craft S,
disease and Down syndrome. Proc Natl Acad Sci USA 82:4245– Haynatzki GR 2006 Rosiglitazone attenuates learning and memory
4249 deficits in Tg2576 Alzheimer mice. Exp Neurol 199:265–273
176. Goedert M 1987 Neuronal localization of amyloid ␤ protein pre- 197. Heneka MT, Sastre M, Dumitrescu-Ozimek L, Hanke A,
cursor mRNA in normal human brain and in Alzheimer’s disease. Dewachter I, Kuiperi C, O’Banion K, Klockgether T, Van Leuven
EMBO J 6:3627–3632 F, Landreth GE 2005 Acute treatment with the PPAR␥ agonist
177. Sinha S, Lieberburg I 1999 Cellular mechanisms of ␤-amyloid pioglitazone and ibuprofen reduces glial inflammation and
production and secretion. Proc Natl Acad Sci USA 96:11049 –11053 A␤1– 42 levels in APPV717I transgenic mice. Brain 128:1442–1453
178. Gasparini L, Gouras GK, Wang R, Gross RS, Beal MF, Greengard 198. Landreth G 2006 PPAR␥ agonists as new therapeutic agents for the
P, Xu H 2001 Stimulation of ␤-amyloid precursor protein trafficking treatment of Alzheimer’s disease. Exp Neurol 199:245–248
by insulin reduces intraneuronal ␤-amyloid and requires mitogen- 199. Inestrosa NC, Godoy JA, Quintanilla RA, Koenig CS, Bronfman
activated protein kinase signaling. J Neurosci 21:2561–2570 M 2005 Peroxisome proliferator-activated receptor ␥ is expressed
179. Vekrellis K, Ye Z, Qiu WQ, Walsh D, Hartley D, Chesneau V, in hippocampal neurons and its activation prevents ␤-amyloid
Rosner MR, Selkoe DJ 2000 Neurons regulate extracellular levels neurodegeneration: role of Wnt signaling. Exp Cell Res 304:91–104
of amyloid ␤-protein via proteolysis by insulin-degrading enzyme. 200. Camacho IE, Serneels L, Spittaels K, Merchiers P, Dominguez D,
J Neurosci 20:1657–1665 De Strooper B 2004 Peroxisome-proliferator-activated receptor ␥
180. Sasaki N, Toki S, Chowei H, Saito T, Nakano N, Hayashi Y, induces a clearance mechanism for the amyloid-␤ peptide. J Neu-
Takeuchi M, Makita Z 2001 Immunohistochemical distribution of rosci 24:10908 –10917
the receptor for advanced glycation end products in neurons and 201. d’Abramo C, Ricciarelli R, Pronzato MA, Davies P 2006 Trogli-
astrocytes in Alzheimer’s disease. Brain Res 888:256 –262 tazone, a peroxisome proliferator-activated receptor-␥ agonist, de-
181. Craft S, Asthana S, Schellenberg G, Baker L, Cherrier M, Boyt creases tau phosphorylation in CHOtau4R cells. J Neurochem 98:
AA, Martins RN, Raskind M, Peskind E, Plymate S 2000 Insulin 1068 –1077
effects on glucose metabolism, memory, and plasma amyloid pre- 202. Lago RM, Singh PP, Nesto RW 2007 Congestive heart failure and
cursor protein in Alzheimer’s disease differ according to apoli- cardiovascular death in patients with prediabetes and type 2 dia-
poprotein-E genotype. Ann NY Acad Sci 903:222–228 betes given thiazolidinediones: a meta-analysis of randomised clin-
182. Zhao L, Teter B, Morihara T, Lim GP, Ambegaokar SS, Ubeda OJ, ical trials. Lancet 370:1129 –1136
Frautschy SA, Cole GM 2004 Insulin-degrading enzyme as a 203. Nissen SE, Wolski K 2007 Effect of rosiglitazone on the risk of
downstream target of insulin receptor signaling cascade: implica- myocardial infarction and death from cardiovascular causes.
tions for Alzheimer’s disease intervention. J Neurosci 24:11120 – N Engl J Med 356:2457–2471
11126 204. Ryan CM 1999 Memory and metabolic control in children. Diabetes
183. Gouras GK, Tsai J, Naslund J, Vincent B, Edgar M, Checler F, Care 22:1239 –1241
Greenfield JP, Haroutunian V, Buxbaum JD, Xu H, Greengard P, 205. Williams AD 2001 Psychometric concerns in neuropsychological
Relkin NR 2000 Intraneuronal A␤42 accumulation in human brain. testing. NeuroRehabilitation 16:221–224
Am J Pathol 156:15–20 206. Walsh P, Kane N, Butler S 2005 The clinical role of evoked po-
184. LaFerla FM, Green KN, Oddo S 2007 Intracellular amyloid-␤ in tentials. J Neurol Neurosurg Psychiatry 76(Suppl 2):ii16 –ii22
Alzheimer’s disease. Nat Rev Neurosci 8:499 –509 207. Dejgaard A, Gade A, Larsson H, Balle V, Parving A, Parving HH
185. Mochizuki A, Tamaoka A, Shimohata A, Komatsuzaki Y, Shoji 1991 Evidence for diabetic encephalopathy. Diabet Med 8:162–167
S 2000 A␤42-positive non-pyramidal neurons around amyloid 208. Martini A, Comacchio F, Fedele D, Crepaldi G, Sala O 1987
plaques in Alzheimer’s disease. Lancet 355:42– 43 Auditory brainstem evoked responses in the clinical evaluation and
186. Westermark P, Wilander E 1978 The influence of amyloid deposits follow-up of insulin-dependent diabetic subjects. Acta Otolaryngol
on the islet volume in maturity onset diabetes mellitus. Diabeto- 103:620 – 627
logia 15:417– 421 209. Parving A, Elberling C, Balle V, Parbo J, Dejgaard A, Parving HH
187. Schwartz P 1965 Senile cerebral, pancreatic insular and cardiac 1990 Hearing disorders in patients with insulin-dependent diabetes
amyloidosis. Trans NY Acad Sci 27:393– 413 mellitus. Audiology 29:113–121
188. Cooper GJ, Willis AC, Clark A, Turner RC, Sim RB, Reid KB 1987 210. Kurita A, Katayama K, Mochio S 1996 Neurophysiological evi-

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 22 May 2014. at 00:09 For personal use only. No other uses without permission. . All rights reserved.
Kodl and Seaquist • Cognition and Diabetes Mellitus Endocrine Reviews, June 2008, 29(4):494 –511 511

dence for altered higher brain functions in NIDDM. Diabetes Care of type 1 diabetes on gray matter density as measured by voxel-
19:360 –364 based morphometry. Diabetes 55:326 –333
211. Gupta PR, Dorfman LJ 1981 Spinal somatosensory conduction in 222. Wessels AM, Simsek S, Remijnse PL, Veltman DJ, Biessels GJ,
diabetes. Neurology 31:841– 845 Barkhof F, Scheltens P, Snoek FJ, Heine RJ, Rombouts SA 2006
212. Pozzessere G, Rizzo PA, Valle E, Mollica MA, Meccia A, Morano Voxel-based morphometry demonstrates reduced grey matter den-
S, Di Mario U, Andreani D, Morocutti C 1988 Early detection of sity on brain MRI in patients with diabetic retinopathy. Diabeto-
neurological involvement in IDDM and NIDDM. Multimodal logia 49:2474 –2480
evoked potentials versus metabolic control. Diabetes Care 11:473– 223. Perantie DC, Wu J, Koller JM, Lim A, Warren SL, Black KJ, Sadler
480 M, White NH, Hershey T 2007 Voxel-based morthometry detects
213. Lobmann R, Smid HG, Pottag G, Wagner K, Heinze HJ, Lehnert brain volume changes due to hypo- and hyperglycemia in youth
H 2000 Impairment and recovery of elementary cognitive function with type 1 diabetes mellitus (TIDM). Diabetes 56(Suppl 1)
induced by hypoglycemia in type-1 diabetic patients and healthy (Abstract 1886)
controls. J Clin Endocrinol Metab 85:2758 –2766 224. Rosenthal JM, Amiel SA, Yaguez L, Bullmore E, Hopkins D,
214. Amiel SA, Pottinger RC, Archibald HR, Chusney G, Cunnah DT, Evans M, Pernet A, Reid H, Giampietro V, Andrew CM, Suckling
Prior PF, Gale EA 1991 Effect of antecedent glucose control on J, Simmons A, Williams SC 2001 The effect of acute hypoglycemia
cerebral function during hypoglycemia. Diabetes Care 14:109 –118 on brain function and activation: a functional magnetic resonance
215. Haumont D, Dorchy H, Pelc S 1979 EEG abnormalities in diabetic imaging study. Diabetes 50:1618 –1626
children: influence of hypoglycemia and vascular complications. 225. Musen G, Bolo N, Jacobson A, Southwick A 2007 Brain functional
correlates of gray matter changes in type 1 diabetes. Diabetes
Clin Pediatr (Phila) 18:750 –753
56(Suppl 1) (Abstract 792)
216. Akisaki T, Sakurai T, Takata T, Umegaki H, Araki A, Mizuno S,
226. Niwa H, Koumoto C, Shiga T, Takeuchi J, Mishima S, Segawa T,
Tanaka S, Ohashi Y, Iguchi A, Yokono K, Ito H 2006 Cognitive
Atsumi T, Shimizu C, Koike T, Yoshioka N 2006 Clinical analysis
dysfunction associates with white matter hyperintensities and sub-
of cognitive function in diabetic patients by MMSE and SPECT.
cortical atrophy on magnetic resonance imaging of the elderly Diabetes Res Clin Pract 72:142–147
diabetes mellitus Japanese elderly diabetes intervention trial 227. Keymeulen B, de Metz K, Cluydts R, Bossuyt A, Somers G 1996
(J-EDIT). Diabetes Metab Res Rev 22:376 –384 Technetium-99m hexamethylpropylene amine oxime single-
217. Yousem DM, Tasman WS, Grossman RI 1991 Proliferative reti- photon emission tomography of regional cerebral blood flow in
nopathy: absence of white matter lesions at MR imaging. Radiology insulin-dependent diabetes. Eur J Nucl Med 23:163–168
179:229 –230 228. Sabri O, Hellwig D, Schreckenberger M, Schneider R, Kaiser HJ,
218. Manschot SM, Brands AM, van der Grond J, Kessels RP, Algra Wagenknecht G, Mull M, Buell U 2000 Influence of diabetes
A, Kappelle LJ, Biessels GJ 2006 Brain magnetic resonance imag- mellitus on regional cerebral glucose metabolism and regional ce-
ing correlates of impaired cognition in patients with type 2 dia- rebral blood flow. Nucl Med Commun 21:19 –29
betes. Diabetes 55:1106 –1113 229. Lim KO, Choi SJ, Pomara N, Wolkin A, Rotrosen JP 2002 Reduced
219. den Heijer T, Vermeer SE, van Dijk EJ, Prins ND, Koudstaal PJ, frontal white matter integrity in cocaine dependence: a controlled
Hofman A, Breteler MM 2003 Type 2 diabetes and atrophy of diffusion tensor imaging study. Biol Psychiatry 51:890 – 895
medial temporal lobe structures on brain MRI. Diabetologia 46: 230. Wozniak JR, Mueller BA, Chang PN, Muetzel RL, Caros L, Lim
1604 –1610 KO 2006 Diffusion tensor imaging in children with fetal alcohol
220. Ferguson SC, Blane A, Wardlaw J, Frier BM, Perros P, McCrimmon spectrum disorders. Alcohol Clin Exp Res 30:1799 –1806
RJ, Deary IJ 2005 Influence of an early-onset age of type 1 diabetes on 231. Wu J, Perantie DC, Sadler M, White NH, Hershey T 2007 Pilot
cerebral structure and cognitive function. Diabetes Care 28:1431–1437 diffusion tensor imaging study in youth with type 1 diabetes mel-
221. Musen G, Lyoo IK, Sparks CR, Weinger K, Hwang J, Ryan CM, litus reveals change in white matter integrity associated with
Jimerson DC, Hennen J, Renshaw PF, Jacobson AM 2006 Effects chronic hyperglycemia. Diabetes 56(Suppl 1) (Abstract 1889)

Endocrine Reviews is published by The Endocrine Society (http://www.endo-society.org), the foremost professional society serving the
endocrine community.

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 22 May 2014. at 00:09 For personal use only. No other uses without permission. . All rights reserved.

You might also like